EP3625251A1 - Anticorps monoclonal anti-virus de la grippe à large neutralisation et utilisations associées - Google Patents

Anticorps monoclonal anti-virus de la grippe à large neutralisation et utilisations associées

Info

Publication number
EP3625251A1
EP3625251A1 EP18732970.1A EP18732970A EP3625251A1 EP 3625251 A1 EP3625251 A1 EP 3625251A1 EP 18732970 A EP18732970 A EP 18732970A EP 3625251 A1 EP3625251 A1 EP 3625251A1
Authority
EP
European Patent Office
Prior art keywords
antibody
antigen
binding
antibodies
binding fragment
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP18732970.1A
Other languages
German (de)
English (en)
Inventor
James J. KOBIE
Michael PIEPENBRINK
Michael KEEFER
Luis MARTINEZ-SOBRIDO
Aitor NOGALES
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Rochester
Original Assignee
University of Rochester
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of Rochester filed Critical University of Rochester
Publication of EP3625251A1 publication Critical patent/EP3625251A1/fr
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/08Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses
    • C07K16/10Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from viruses from RNA viruses
    • C07K16/1018Orthomyxoviridae, e.g. influenza virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16111Influenzavirus A, i.e. influenza A virus
    • C12N2760/16134Use of virus or viral component as vaccine, e.g. live-attenuated or inactivated virus, VLP, viral protein

Definitions

  • This invention relates to broadly neutralizing anti-influenza monoclonal antibodies (mAbs) or antigen-binding fragments thereof.
  • the present invention further relates to the therapeutic uses of the antibody or the antigen-binding fragment.
  • Influenza commonly known as "the flu” is an infectious disease caused by influenza virus.
  • influenza viruses There are four types of influenza viruses: A, B, C and D.
  • Human influenza A and B viruses cause seasonal epidemics of the disease.
  • the first and most important step in preventing flu is to get an annual flu vaccination.
  • a licensed influenza vaccine has been available for over seventy years, influenza infections still remain a major public health concern.
  • influenza leads to -15,000 deaths and -300,000 hospitalizations, with -3 to 5 million severe cases and 200,000 to 500,000 deaths per year globally (Girard MP, et al. 2005. Vaccine 23 :5708-5724; Nogales A, et al. 2016. Int J Mol Sci 18; Dushoff J, et al. 2006.
  • Influenza A virus has 18 HA subtypes, which are further classified in two phylogenetic groups: group 1 (HI, H2, H5, H6, H8, H9, HI 1, H12, H13, H16, H17 and H18 subtypes) and group 2 (H3, H4, H7, H10, H14 and H15 subtypes).
  • Seasonal vaccinations include influenza type A HI, H3, and type B viruses.
  • Recent pandemics including the latest 2009 novel HlNl pandemic (Smith GJ, et al. 2009. Nature 459: 1122-1 125 and Kilbourne ED. 2006.
  • This invention addresses the need by providing broadly neutralizing anti-influenza monoclonal antibodies or antigen-binding fragments thereof.
  • the invention provides an isolated antibody or antigen-binding fragment thereof that specifically binds to a hemagglutinin (HA) of influenza A virus (IAV) HI subtype, comprising: (i) a heavy chain variable region that comprises HCDR1, HCDR2, and HCDR3 comprising the amino acid sequences of SEQ ID NOs: 3-5, respectively, and (ii) a light chain variable region that comprises LCDR1, LCDR2 and LCDR3 comprising the amino acid sequences of SEQ ID NOs: 6-8, respectively.
  • the heavy chain variable region can include the amino acid sequence of SEQ ID NO: 1.
  • the light chain variable region can include the amino acid sequence of SEQ ID NO: 2.
  • the invention also provides an isolated antibody or antigen-binding fragment thereof that specifically binds to an HA of IAV HI subtype. When bound to the HA, the antibody binds to a conformational epitope dependent on (or containing) the E and K amino acid residues corresponding to E129 and K180 of the HA of pHlNl (SEQ ID NO: 13). Further provided is an isolated antibody or the antigen-binding fragment thereof that competes for binding to an HA of IAV HI subtype in a cross-blocking assay with the antibody or the antigen-binding fragment described above.
  • the above-described antibody or antigen-binding fragment can include a variant Fc constant region.
  • the isolated antibody or the antigen-binding fragment can be a chimeric antibody, a humanized antibody, or a human antibody.
  • the antibody or fragment can be conjugated to a therapeutic agent, a polymer, a detectable label, or an enzyme.
  • the polymer include polyethylene glycol (PEG).
  • the therapeutic agent include a cytotoxic agent.
  • the invention provides an isolated nucleic acid encoding one or more of the CDRs, the heavy or light chain variable region, or antigen binding portion, of any one of above-described antibodies or antigen-binding fragments.
  • the nucleic acid can be used to express a polypeptide having one or both sets of the HCDRs or LCDRS, a chain of the antibody or antigen-binding fragment, or the antibody or fragment described above.
  • one can operatively link the nucleic acid to suitable regulatory sequences to generate an expression vector.
  • a cultured host cell comprising the vector and a method for producing a polypeptide, an antibody, or antigen binding portion thereof.
  • the method includes: obtaining a cultured host cell comprising a vector comprising a nucleic acid sequence encoding one or more of the above mentioned CDRs, polypeptide, a heavy chain variable region or a light chain variable region of the antibody or antigen binding portion thereof as described above; culturing the cell in a medium under conditions permitting expression of a polypeptide encoded by the vector and assembling of an antibody or fragment thereof, and purifying the antibody or fragment from the cultured cell or the medium of the cell.
  • the antibody or fragment described above can be used in a method of neutralizing IAV or a method of treating, preventing or controlling an IAV infection.
  • the method includes administering to a subject in need thereof a therapeutically effective amount of the antibody or fragment.
  • the invention also provides a pharmaceutical composition comprising (i) the antibody or an antigen-binding fragment thereof, and (ii) a pharmaceutically acceptable carrier.
  • FIGs. 1A, IB, and 1C are a set of diagrams showing isolation and molecular characterization of KPF1 human monoclonal antibody (hmAb).
  • FIG. 1A Gating strategy to isolate peripheral blood plasmablasts (CD19+IgD-CD38+CD27++) 7 days after immunization.
  • FIG. IB Alignment of KPFl VH and Vk (SEQ ID NOs: 1 and 9) with presumed germline amino acid sequences (SEQ ID NOs: 10 and 11).
  • FIG. 1C Alignment of nucleic acids encoding a constant region of an antibody from an expanded B cell lineage (SEQ ID NO: 22) used to make KPF 1 and an IgAl constant region (SEQ ID NO: 23).
  • FIGs. 2 A, 2B and 2C are diagrams showing that KPFl hmAb is highly specific for HI influenza.
  • KPFl hmAb and isotype control hmAb were tested by ELISA for binding to (FIG. 2 A) diverse recombinant influenza HI, H3, H5, H7, H9 and IBV HAs and negative control protein (RSV-F) and (FIG. 2B) HI HA proteins in increasing concentrations of urea. Symbols represent triplicate ⁇ SEM.
  • FIG. 2C Purified KPFl was captured on a Protein G chip with the pHlNl HA at decreasing concentrations passed over each channel. The data points are shown in black and the fit to a 1 : 1 binding model are shown in red. The results of one representative experiments of two are presented.
  • FIG. 3 is a diagram showing mPLEX-Flu binding profile.
  • the patient's plasma from before (DO) and after 7 days (D7) and 3 months (M3) post immunization, and KPFl hmAb were tested in decreasing concentration by multiplex assay for binding to diverse recombinant HI, H2, H3, H5, H6, H7, H9 and IBV HA proteins.
  • FIGs. 4A, 4B, and 4C are diagrams and a table showing potent in vitro neutralizing activity of KPFl hmAb.
  • Virus neutralization was measured using a fluorescent-based microneutralization assay (Nogales A, et al. 2016. Virus Res 213 :69-81 ; and Nogales A, et al. 2015. Virology 476:206-216).
  • MDCK cells were infected with mCherry-expressing A/California/04/2009 (pHlNl) and A/Puerto Rico/08/1934 (PR8) H1N1, A/Wyoming/3/2003 (H3N2), or B/Brisbane/60/2008 (IBV) viruses, which were pre-incubated with two-fold serial dilutions of KPFl hmAb.
  • virus neutralization was evaluated and quantified using a fluorescence microplate reader (FIG. 4A), and the percentage of infectivity calculated using sigmoidal dose response curves (FIG. 4B).
  • FIGs. 5 A, 5B, 5C and 5D are diagrams showing that KPFl hmAb restricts pHlNl replication in vivo.
  • To evaluate viral replication in the lungs FIG.
  • Female C57BL/6 mice (N 6) received 10 mg/kg of KPF 1 or IgG isotype control (IC) 24 h before viral infection.
  • symbols represent data from individual mice. Bars, geometric mean lung virus titers; dotted line, limit of detection (200 FFU/ml). & indicates virus was not detected or was detected only in 1 of 3 mice.
  • FIGs. 6A and 6B are diagrams showing therapeutic activity of KPF1 in infected mice.
  • FIGs. 7A, 7B, 7C, 7D and 7E are tables, a set of photographs, and diagrams showing generation and characterization of MARMs.
  • FIG. 7A Amino acid mutations in the HAs and NAs of WT or mAb-resistant mutants (MARMs 1, 2 and 3) after 5 rounds of selection in the presence (MARMs) or absence (WT) of hmAb KPF1. The mutations effects on reactivity with the hmAb KPF 1 were also evaluated in a microneutralization assay (NT50) and HAL
  • FIG. 7B Characterization of MARMs by immunofluorescence.
  • MDCK cells were mock infected (Mock) or infected (MOI 0.01) with pHlNl WT or the MARMs (1, 2 and 3). At 36 h p.i., cells were fixed and protein expression was evaluated by IFA using the hmAb KPF1, or the mouse mAbs 29E3 (anti-HA) and HB-65 (anti-NP). DAPI was used for nuclear staining. Merge from representative images (1 OX magnification) are included. Scale bar, 50 nm. (FIG. 7C) Multicycle growth kinetics of pHlNl WT and MARMs in MDCK cells.
  • Virus titers in tissue culture supernatants of MDCK cells infected (MOI, 0.001) with pHlNl WT or MARMs viruses were analyzed at the indicated h p.i by immunofocus assay (FFU/ml) using the anti-NP mouse mAb HB-65. Data represent the means ⁇ SDs of the results determined for triplicate wells. * indicates p ⁇ 0.05 (WT versus MARM 3) using a Student's t test.
  • FIG. 7D Tridimensional protein structure for the globular head of HA of pHlNl . The image was created using the software program PyMol and the published structure for the HA of pHlNl (3LZG, Xu R, et al. 2010.
  • FIGs. 8A and 8B are a set of photographs and diagrams showing relevance of amino acids 129 and 180 for the binding of KPF1 hmAb.
  • 8A Binding of KPF1 hmAb to WT and mutant HA proteins.
  • HEK293T cells were transiently transfected with the pCAGGS plasmids expressing WT or amino acid substitutions E129K, K180N, K180Q or E129K/K180N mutant HAs. Mock transfected cells were used as internal control.
  • cells were fixed and protein expression was evaluated by IF A using the hmAb KPF1, or a goat pHlNl anti-HA polyclonal antibody as a control.
  • DAPI was used for nuclear staining. Merge from representative images (lOx magnification) are included. Scale bar, 50 nm.
  • This invention is based, at least in part, on unexpected broadly neutralizing anti- influenza activities of certain monoclonal antibodies or antigen-binding fragments thereof. These antibodies and antigen-binding fragments constitute a novel therapeutic strategy in protection from influenza infections.
  • the invention disclosed herein involves broadly neutralizing anti-influenza monoclonal antibodies or antigen-binding fragments thereof.
  • These antibodies refer to a class of neutralizing antibodies that neutralize multiple influenza virus strains.
  • the antibodies are able to protect prophylactically and therapeutically a subject (e.g., a mouse as shown in the examples below) against a lethal challenge with an influenza virus, such as A/California/04/2009 H1N1 (pHlNl).
  • Each of the antibodies binds to a conserved epitope region of the HA globular head near the receptor binding site (RBS) different than that previously described to other cross-reactive HI mAbs.
  • RBS receptor binding site
  • these antibodies recognize a highly conserved, novel discontinuous (or conformational) epitope in the HA1 globular head of HI influenza strains that dependent on residues within the Sa antigenic site (K180) and near the Ca antigenic site (E129), encompassing a region near the RBS.
  • hmAbs human monoclonal antibodies
  • the neutralization titer 50 (NT50) of the antibody can be as low as less than 10.0, 5.0, 1.0, 0.5, 0.10, 0.05, 0.04, 0.03, or 0.02 ⁇ g/ml. Most of the binding (-50-70%) to HA can be maintained even in 8M Urea indicating high avidity.
  • an antibody of this invention can recognize a large number of HI isolates, including one, two, three, four, or five of A/California/07/2009 H1N1, A/New Caledonia/20/1999 H1N1, A/Texas/36/1991 H1N1, A/South Carolina/01/1918 H1N1, and A/Puerto Rico/08/1934 H1N1.
  • KPFl recognized 83% of all HI isolates tested, including 1918 HI .
  • the antibody does not recognize A/USSR/1977 H1N1 at 1 ⁇ g/ml, od has no reactivity against H3 or B HAs.
  • the antibody of this invention ⁇ e.g., KPFl
  • a high percentage e.g., 50%, 60%, 70%, 80%, 90%, 95% or 100%
  • an antibody of this invention ⁇ e.g., KPFl
  • a high percentage e.g., 40%, 50%, 60%, 70%, or 80%
  • the antibody of this invention ⁇ e.g., KPFl
  • KPFl recognizes an epitope in the HA globular head that is dependent on residues within the Sa antigenic site and near the receptor binding site.
  • an antibody of this invention (such as KPFl) has more potent neutralizing anti-influenza activities.
  • HC heavy chain variable regions
  • LC light chain variable regions of one exemplary antibody, the KPFl antibody mentioned above, where the heavy chain CDRl-3 (HCDRl, HCDR2, and HCDR3) and light chain CDR1-3(LCDR1, LCDR2, and LCDR3) are in bold.
  • HA hemagglutinin
  • H1N1 A/California/07/2009 X-179A
  • H3N2 A/Texas/50/2012 X-223A
  • B/Massachusetts/02/2012 B Yamagata lineage
  • B/Brisbane/60/2008 Victoria lineage
  • caattgagct cagtgtcatc atttgaaagg tttgagatat tccccaagac aagttcatgg
  • nucleotide sequence(SEQ ID NO : 20 ) Shown below are the nucleotide and the amino acid sequence of the pHlNl HA. Nucleotide sequence(SEQ ID NO : 20 ) :
  • Amino acid sequence (SEQ ID NO : 21 ) :
  • Antibodies disclosed herein have broad activity against HI influenza isolates and potent prophylactic and therapeutic activity / « v/ ' vo, which is mediated by recognition of conserved residues in the HI hemagglutinin globular head.
  • Each of the antibodies is highly specific to influenza HI HA and recognizes all HI isolates tested with the exception of A/USSR/1977, likely due to the unique HA structure of this pandemic isolate (Kilbourne ED. 2006. Emerg Infect Dis 12:9-14; and Rozo M, et al. 2015. MBio 6).
  • the high potency of the antibody is demonstrated in vitro by its neutralizing and HAI activities below 1.0 ⁇ g/ml (Fig. 4), its ability to maintain HA binding in the presence of urea, and its high avidity and affinity (Fig. 2).
  • Few hmAbs have been reported that have similar in vitro neutralizing activity of HI influenza below 1 ⁇ g/ml (Sparrow E, et al. 2016. Vaccine 34:5442-5448; Whittle JR, et al. 2011. Proc Natl Acad Sci U S A 108: 14216-14221; Krause JC, et al. 2011. J Virol 85: 10905-10908; Ren H, et al. 2016.
  • the potent activity of the antibody of this invention extends to its ability to protect and treat HI infection in vivo (Figs. 5 and 6).
  • the challenge dose of lOx MLD50 pHlNl used in the study shown below exceeded by 2 to 5 fold that which has been used by others (Heaton NS, et al. 2013. J Virol 87:8272-8281 ; Wang SF, et al. 2016. Dev Comp Immunol doi: 10.1016/j dci.2016.10.010; Marjuki H, et al. 2016. J Virol 90: 10446-10458; Song A, et al. 2014. Antiviral Res 111 :60-68; DiLillo DJ, et al.
  • the antibody of this invention recognizes a highly conserved novel epitope (Fig. 8) in the HA1 globular head of HI influenza strains that dependent on the E129 residue near the Ca and Cb antigenic sites (Fig. 7).
  • the hmAb 2D1 which recognizes both 1918 and pHlNl HA1, but has limited activity against most influenza HI strains, recognizes an epitope centered on Sa which includes K180 (Xu R, et al. 2010. Science 328:357-360), and selected for escape mutants at this residue (Krause JC, et al. 2010. J Virol 84:3127- 3130).
  • 2D1 was not reported to interact with El 29, suggesting the precise epitopes recognized by the antibody of this invention, such as KPF1, are distinct than those of 2D1.
  • the mouse mAb GC0587 which was generated from pHlNl immunized mice is Hl-specific and recognizes an epitope that contains both E129 and K180 residues (Cho KJ, et al. 2014. PLoS One 9:e89803). However, its lack of reactivity against 1918 HI (Cho KJ, et al. 2014. PLoS One 9:e89803) in contrast to the antibody of this invention, such as KPF1, suggesting incomplete congruence in the epitopes recognized by both mAbs.
  • ADCC Ab-dependent cell-mediated cytotoxicity
  • CDC complement-dependent cytotoxicity
  • KPF1 as well as the E129 amino acid conservation, suggests it has therapeutic value for the treatment and prevention of HI influenza infections.
  • hmAbs targeting HA stem epitopes have been identified that have broadly neutralizing activity against multiple influenza types and subtypes, their potency is commonly less than hmAbs targeting the HA globular head, and their clinical efficacy has yet to be determined.
  • influenza viruses causing pandemics have been subtype specific ⁇ e.g. H1N1, H2N2 or H3N2) and, therefore, having more potent globular head broadly neutralizing antibodies rather than less specific and less potent neutralizing stalk reactive antibodies represents a better approach for pandemic preparedness.
  • a cocktail of multiple high affinity type-specific hmAbs including the antibody of this invention, such as KPF1
  • an antibody provided herein is an antibody fragment.
  • Antibody fragments include, but are not limited to, Fab, Fab', Fab'-SH, F(ab') 2 , Fv, and scFv fragments, and other fragments described below, e.g., diabodies. triabodies tetrabodies, and single-domain antibodies.
  • Fab fragment antigen
  • Fab' fragment antigen binding domain
  • Fab'-SH fragment antigen binding
  • F(ab') 2 fragment antigen Vv fragment fragment
  • Fv fragment antigen fragment fragment fragments
  • scFv fragments see, e.g., Pluckthun, in The Pharmacology of Monoclonal Antibodies, vol.
  • Diabodies are antibody fragments with two antigen-binding sites that may be bivalent or bispecific. See, for example, EP 404,097; WO 1993/01161; Hudson et al, Nat. Med. 9: 129-134 (2003); and Hollinger et al, Proc. Natl. Acad. Sci. USA 90: 6444-6448 (1993). Triabodies and tetrabodies are also described in Hudson et al, Nat. Med. 9: 129-134 (2003).
  • Single-domain antibodies are antibody fragments comprising all or a portion of the heavy chain variable domain or all or a portion of the light chain variable domain of an antibody.
  • a single-domain antibody is a human single-domain antibody (Domantis, Inc., Waltham, Mass.; see, e.g., U.S. Pat. No. 6,248,516).
  • Antibody fragments can be made by various techniques, including but not limited to proteolytic digestion of an intact antibody as well as production by recombinant host cells (e.g., E. coli or phage), as described herein.
  • recombinant host cells e.g., E. coli or phage
  • an antibody provided herein is a chimeric antibody.
  • Certain chimeric antibodies are described, e.g., in U.S. Pat. No. 4,816,567; and Morrison et al., Proc. Natl. Acad. Sci. USA, 81 :6851-6855 (1984)).
  • a chimeric antibody comprises a non-human variable region ⁇ e.g., a variable region derived from a mouse, rat, hamster, rabbit, or non-human primate, such as a monkey) and a human constant region.
  • a chimeric antibody is a "class switched" antibody in which the class or subclass has been changed from that of the parent antibody. Chimeric antibodies include antigen-binding fragments thereof.
  • a chimeric antibody is a humanized antibody.
  • a non-human antibody is humanized to reduce immunogenicity to humans, while retaining the specificity and affinity of the parental non-human antibody.
  • a humanized antibody comprises one or more variable domains in which HVRs, e.g., CDRs, (or portions thereof) are derived from a non-human antibody, and FRs (or portions thereof) are derived from human antibody sequences.
  • HVRs e.g., CDRs, (or portions thereof) are derived from a non-human antibody
  • FRs or portions thereof
  • a humanized antibody optionally will also comprise at least a portion of a human constant region.
  • some FR residues in a humanized antibody are substituted with corresponding residues from a non-human antibody (e.g., the antibody from which the HVR residues are derived), e.g., to restore or improve antibody specificity or affinity.
  • a non-human antibody e.g., the antibody from which the HVR residues are derived
  • Human framework regions that may be used for humanization include but are not limited to: framework regions selected using the "best-fit" method (see, e.g., Sims et al. J. Immunol. 151 :2296 (1993)); framework regions derived from the consensus sequence of human antibodies of a particular subgroup of light or heavy chain variable regions (see, e.g., Carter et al. Proc. Natl. Acad. Sci. USA, 89:4285 (1992); and Presta et al. J. Immunol., 151 :2623 (1993)); human mature (somatically mutated) framework regions or human germline framework regions (see, e.g., Almagro and Fransson, Front. Biosci.
  • an antibody provided herein is a human antibody.
  • Human antibodies can be produced using various techniques known in the art or using techniques described herein. Human antibodies are described generally in van Dijk and van de Winkel, Curr. Opin. Pharmacol. 5: 368-74 (2001) and Lonberg, Curr. Opin. Immunol. 20:450-459 (2008).
  • Human antibodies may be prepared by administering an immunogen to a transgenic animal that has been modified to produce intact human antibodies or intact antibodies with human variable regions in response to antigenic challenge.
  • Such animals typically contain all or a portion of the human immunoglobulin loci, which replace the endogenous immunoglobulin loci, or which are present extrachromosomally or integrated randomly into the animal's chromosomes.
  • the endogenous immunoglobulin loci have generally been inactivated.
  • Human antibodies can also be made by hybridoma-based methods. Human myeloma and mouse-human heteromyeloma cell lines for the production of human monoclonal antibodies have been described. (See, e.g., Kozbor J. Immunol., 133 : 3001 (1984); Brodeur et al, Monoclonal Antibody Production Techniques and Applications, pp. 51-63 (Marcel Dekker, Inc., New York, 1987); and Boerner et al, J. Immunol., 147: 86 (1991).) Human antibodies generated via human B-cell hybridoma technology are also described in Li et al., Proc. Natl. Acad. Sci. USA, 103 :3557-3562 (2006).
  • Additional methods include those described, for example, in U.S. Pat. No. 7, 189,826 (describing production of monoclonal human IgM antibodies from hybridoma cell lines) and Ni, Xiandai Mianyixue, 26(4):265-268 (2006) (describing human-human hybridomas).
  • Human hybridoma technology Trioma technology
  • Vollmers and Brandlein, Histology and Histopathology, 20(3):927-937 (2005) and Vollmers and Brandlein, Methods and Findings in Experimental and Clinical Pharmacology, 27(3): 185-91 (2005).
  • Human antibodies may also be generated by isolating Fv clone variable domain sequences selected from human-derived phage display libraries. Such variable domain sequences may then be combined with a desired human constant domain. Techniques for selecting human antibodies from antibody libraries are described below.
  • Antibodies of the invention may be isolated by screening combinatorial libraries for antibodies with the desired activity or activities. For example, a variety of methods are known in the art for generating phage display libraries and screening such libraries for antibodies possessing the desired binding characteristics. Such methods are reviewed, e.g., in Hoogenboom et al. in Methods in Molecular Biology 178: 1-37 (O'Brien et al., ed., Human Press, Totowa, N.J., 2001) and further described, e.g., in the McCafferty et al., Nature 348:552-554; Clackson et al, Nature 352: 624-628 (1991); Marks et al, J. Mol. Biol.
  • repertoires of VH and VL genes are separately cloned by polymerase chain reaction (PCR) and recombined randomly in phage libraries, which can then be screened for antigen-binding phage as described in Winter et al, Ann. Rev. Immunol., 12: 433-455 (1994).
  • Phage typically display antibody fragments, either as single-chain Fv (scFv) fragments or as Fab fragments.
  • naive repertoire can be cloned ⁇ e.g., from human) to provide a single source of antibodies to a wide range of non-self and also self antigens without any immunization as described by Griffiths et al, EMBO J, 12: 725-734 (1993).
  • naive libraries can also be made synthetically by cloning unrearranged V-gene segments from stem cells, and using PCR primers containing random sequence to encode the highly variable CDR3 regions and to accomplish rearrangement in vitro, as described by Hoogenboom and Winter, J. Mol. Biol., 227: 381-388 (1992).
  • Patent publications describing human antibody phage libraries include, for example: U.S. Pat. No.
  • Antibodies or antibody fragments isolated from human antibody libraries are considered human antibodies or human antibody fragments herein.
  • amino acid sequence variants of the antibodies provided herein are contemplated. For example, it may be desirable to improve the binding affinity and/or other biological properties of the antibody.
  • Amino acid sequence variants of an antibody may be prepared by introducing appropriate modifications into the nucleotide sequence encoding the antibody, or by peptide synthesis. Such modifications include, for example, deletions from, and/or insertions into and/or substitutions of residues within the amino acid sequences of the antibody. Any combination of deletion, insertion, and substitution can be made to arrive at the final construct, provided that the final construct possesses the desired characteristics, e.g., antigen-binding.
  • antibody variants having one or more amino acid substitutions are provided.
  • Sites of interest for substitutional mutagenesis include the HVRs and FRs.
  • Conservative substitutions are defined herein.
  • Amino acid substitutions may be introduced into an antibody of interest and the products screened for a desired activity, e.g., retained/improved antigen binding, decreased immunogenicity, or improved ADCC or CDC.
  • an antibody of the invention can comprise one or more conservative modifications of the CDRs, heavy chain variable region, or light variable regions described herein, e.g., SEQ ID NOs: 1-8.
  • a conservative modification or functional equivalent of a peptide, polypeptide, or protein disclosed in this invention refers to a polypeptide derivative of the peptide, polypeptide, or protein, e.g., a protein having one or more point mutations, insertions, deletions, truncations, a fusion protein, or a combination thereof. It retains substantially the activity to of the parent peptide, polypeptide, or protein (such as those disclosed in this invention).
  • a conservative modification or functional equivalent is at least 60% (e.g., any number between 60% and 100%, inclusive, e.g., 60%, 10%, 15%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, and 99%) identical to a parent (e.g., one of SEQ ID NOs: 1-8). Accordingly, within scope of this invention are heavy chain variable region or light variable regions having one or more point mutations, insertions, deletions, truncations, a fusion protein, or a combination thereof, as well as antibodies having the variant regions.
  • the percent homology between two amino acid sequences is equivalent to the percent identity between the two sequences.
  • the comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm, as described in the non-limiting examples below.
  • the percent identity between two amino acid sequences can be determined using the algorithm of E. Meyers and W. Miller (Comput. Appl. Biosci., 4: 11-17 (1988)) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4.
  • the percent identity between two amino acid sequences can be determined using the Needleman and Wunsch (J. Mol. Biol.
  • the protein sequences of the present invention can further be used as a "query sequence" to perform a search against public databases to, for example, identify related sequences. Such searches can be performed using the XBLAST program (version 2.0) of Altschul, et al. (1990) J. Mol. Biol. 215:403-10.
  • Gapped BLAST can be utilized as described in Altschul et al, (1997) Nucleic Acids Res. 25(17):3389-3402.
  • the default parameters of the respective programs ⁇ e.g., XBLAST and NBLAST) can be used. (See www.ncbi.nlm.nih.gov).
  • conservative modifications refers to amino acid modifications that do not significantly affect or alter the binding characteristics of the antibody containing the amino acid sequence. Such conservative modifications include amino acid substitutions, additions and deletions. Modifications can be introduced into an antibody of the invention by standard techniques known in the art, such as site-directed mutagenesis and PCR-mediated mutagenesis. Conservative amino acid substitutions are ones in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art. These families include:
  • amino acids with basic side chains ⁇ e.g., lysine, arginine, histidine
  • acidic side chains ⁇ e.g., aspartic acid, glutamic acid
  • uncharged polar side chains ⁇ e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan
  • nonpolar side chains ⁇ e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine
  • beta-branched side chains ⁇ e.g., threonine, valine, isoleucine
  • aromatic side chains ⁇ e.g., tyrosine, phenylalanine, tryptophan, histidine).
  • Non-conservative substitutions will entail exchanging a member of one of these classes for another class.
  • An exemplary substitutional variant is an affinity matured antibody, which may be conveniently generated, e.g., using phage display -based affinity maturation techniques such as those described in e.g., Hoogenboom et al., in Methods in Molecular Biology 178: 1-37 (O'Brien et al., ed., Human Press, Totowa, N.J., (2001).
  • Amino acid sequence insertions include amino- and/or carboxyl-terminal fusions ranging in length from one residue to polypeptides containing a hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues. Examples of terminal insertions include an antibody with an N-terminal methionyl residue.
  • Other insertional variants of the antibody molecule include the fusion to the N- or C-terminus of the antibody to an enzyme (e.g., for ADEPT) or a polypeptide which increases the serum half-life of the antibody.
  • an antibody provided herein is altered to increase or decrease the extent to which the antibody is glycosylated.
  • Addition or deletion of glycosylation sites to an antibody may be conveniently accomplished by altering the amino acid sequence such that one or more glycosylation sites is created or removed.
  • an aglycoslated antibody can be made (i.e., the antibody lacks glycosylation).
  • Glycosylation can be altered to, for example, increase the affinity of the antibody for antigen.
  • Such carbohydrate modifications can be accomplished by, for example, altering one or more sites of glycosylation within the antibody sequence.
  • one or more amino acid substitutions can be made that result in elimination of one or more variable region framework glycosylation sites to thereby eliminate glycosylation at that site.
  • Such aglycosylation may increase the affinity of the antibody for antigen.
  • Glycosylation of the constant region on N297 may be prevented by mutating the N297 residue to another residue, e.g., N297A, and/or by mutating an adjacent amino acid, e.g., 298 to thereby reduce glycosylation on N297.
  • an antibody can be made that has an altered type of glycosylation, such as a hypofucosylated antibody having reduced amounts of fucosyl residues or an antibody having increased bisecting GlcNac structures.
  • altered glycosylation patterns have been demonstrated to increase the ADCC ability of antibodies.
  • carbohydrate modifications can be accomplished by, for example, expressing the antibody in a host cell with altered glycosylation machinery. Cells with altered glycosylation machinery have been described in the art and can be used as host cells in which to express recombinant antibodies described herein to thereby produce an antibody with altered glycosylation.
  • PCT Publication WO 03/035835 by Presta describes a variant CHO cell line, Led 3 cells, with reduced ability to attach fucose to Asn(297)-linked carbohydrates, also resulting in hypofucosylation of antibodies expressed in that host cell (see also Shields, R.L. et al. (2002) J. Biol. Chem. 277:26733-26740).
  • PCT Publication WO 99/54342 by Umana et al.
  • glycoprotein-modifying glycosyl transferases ⁇ e.g., beta(l,4)-N-acetylglucosaminyltransf erase III (GnTIII)
  • GnTIII glycoprotein-modifying glycosyl transferases
  • variable regions of the antibody described herein can be linked ⁇ e.g., covalently linked or fused) to an Fc, e.g., an IgGl, IgG2, IgG3 or IgG4 Fc, which may be of any allotype or isoallotype, e.g., for IgGl: Glm, Glml(a), Glm2(x), Glm3(f), Glml7(z); for IgG2: G2m, G2m23(n); for IgG3 : G3m, G3m21(gl), G3m28(g5), G3ml l(b0), G3m5(bl), G3ml3(b3), G3ml4(b4), G3ml0(b5), G3ml5(s), G3ml6(t), G3m6(c3), G3m24(c5), G3m26(u), G3m27(v);
  • the antibodies variable regions described herein are linked to an Fc that binds to one or more activating Fc receptors (Fcyl, Fcylla or Fcyllla), and thereby stimulate ADCC and may cause T cell depletion. In certain embodiments, the antibody variable regions described herein are linked to an Fc that causes depletion.
  • the antibody variable regions described herein may be linked to an Fc comprising one or more modification, typically to alter one or more functional properties of the antibody, such as serum half-life, complement fixation, Fc receptor binding, and/or antigen-dependent cellular cytotoxicity.
  • an antibody described herein may be chemically modified ⁇ e.g., one or more chemical moieties can be attached to the antibody) or be modified to alter its glycosylation, to alter one or more functional properties of the antibody.
  • the numbering of residues in the Fc region is that of the EU index of Kabat.
  • the Fc region encompasses domains derived from the constant region of an immunoglobulin, preferably a human immunoglobulin, including a fragment, analog, variant, mutant or derivative of the constant region.
  • Suitable immunoglobulins include IgG 1 , IgG2, IgG3, IgG4, and other classes such as IgA, IgD, IgE and IgM,
  • the constant region of an immunoglobulin is defined as a naturally- occurring or synthetically-produced polypeptide homologous to the immunoglobulin C-terminal region, and can include a CH I domain, a hinge, a CH2 domain, a CH3 domain, or a CH4 domain, separately or in combination.
  • an antibody of this invention has an Fc region other than that of a wild type IgAl .
  • the antibody can have an Fc region from that of IgG (e.g., IgGl, IgG2, IgG3, and IgG4) or other classes such as IgA2, IgD, IgE and IgM.
  • the Fc can be a mutant form of IgAl .
  • the constant region of an immunoglobulin is responsible for many important antibody functions including Fc receptor (FcR) binding and complement fixation.
  • FcR Fc receptor
  • IgG is separated into four subclasses known as IgGl, IgG2, IgG3, and IgG4,
  • Ig molecules interact with multiple classes of cellular receptors.
  • IgG molecules interact with three classes of Fey receptors (FcyR) specific for the IgG class of antibody, namely FcyRI, FcyRII, and FcyRIIL.
  • FcyR Fey receptors
  • the important sequences for the binding of IgG to the FcyR receptors have been reported to be located in the CH2 and CH3 domains.
  • the serum half-life of an antibody is influenced by the ability of that antibody to bind to an Fc receptor (FcR).
  • the Fc region is a variant Fc region, e.g., an Fc sequence that has been modified (e.g., by amino acid substitution, deletion and/or insertion) relative to a parent Fc sequence (e.g., an unmodified Fc polypeptide that is subsequently modified to generate a variant), to provide desirable structural features and/or biological activity.
  • a variant Fc region e.g., an Fc sequence that has been modified (e.g., by amino acid substitution, deletion and/or insertion) relative to a parent Fc sequence (e.g., an unmodified Fc polypeptide that is subsequently modified to generate a variant), to provide desirable structural features and/or biological activity.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • CDC complement mediated cytotoxicity
  • c has increased or decreased affinity for Clq
  • d has increased or decreased affinity for a Fc receptor relative to the parent Fc.
  • Such Fc region variants will generally comprise at least one amino acid modification in the Fc region. Combining amino acid modifications is thought to be particularly desirable.
  • the variant Fc region may include two, three, four, five, etc. substitutions therein, e.g. of the specific Fc region positions identified herein.
  • a variant Fc region may also comprise a sequence alteration wherein amino acids involved in disulfide bond formation are removed or replaced with other amino acids. Such removal may avoid reaction with other cysteine-containing proteins present in the host cell used to produce the antibodies described herein. Even when cysteine residues are removed, single chain Fc domains can still form a dimeric Fc domain that is held together non- covalently.
  • the Fc region may be modified to make it more compatible with a selected host cell. For example, one may remove the PA sequence near the N-terminus of a typical native Fc region, which may be recognized by a digestive enzyme in E. coli such as proline iminopeptidase.
  • one or more glycosylation sites within the Fc domain may be removed. Residues that are typically glycosylated (e.g., asparagine) may confer cytolytic response. Such residues may be deleted or substituted with unglycosylated residues (e.g., alanine).
  • sites involved in interaction with complement such as the Clq binding site, may be removed from the Fc region. For example, one may delete or substitute the EKK sequence of human IgGl.
  • sites that affect binding to Fc receptors may be removed, preferably sites other than salvage receptor binding sites.
  • an Fc region may be modified to remove an ADCC site.
  • ADCC sites are known in the art; see, for example, Molec. Immunol. 29 (5): 633-9 (1992) with regard to ADCC sites in IgGl. Specific examples of variant Fc domains are disclosed for example, in WO 97/34631 and WO 96/32478.
  • the hinge region of Fc is modified such that the number of cysteine residues in the hinge region is altered, e.g., increased or decreased.
  • the number of cysteine residues in the hinge region of Fc is altered to, for example, facilitate assembly of the light and heavy chains or to increase or decrease the stability of the antibody.
  • the Fc hinge region of an antibody is mutated to decrease the biological half-life of the antibody.
  • one or more amino acid mutations are introduced into the CH2- CH3 domain interface region of the Fc-hinge fragment such that the antibody has impaired Staphylococcyl protein A (SpA) binding relative to native Fc-hinge domain SpA binding.
  • SpA Staphylococcyl protein A
  • the Fc region is altered by replacing at least one amino acid residue with a different amino acid residue to alter the effector function(s) of the antibody.
  • one or more amino acids selected from amino acid residues 234, 235, 236, 237, 297, 318, 320 and 322 can be replaced with a different amino acid residue such that the antibody has an altered affinity for an effector ligand but retains the antigen-binding ability of the parent antibody.
  • the effector ligand to which affinity is altered can be, for example, an Fc receptor or the CI component of complement. This approach is described in further detail in U.S. Patent Nos. 5,624,821 and 5,648,260, both by Winter et al.
  • 331 and 322 can be replaced with a different amino acid residue such that the antibody has altered Clq binding and/or reduced or abolished complement dependent cytotoxicity (CDC).
  • CDC complement dependent cytotoxicity
  • the Fc region may be modified to increase antibody dependent cellular cytotoxicity (ADCC) and/or to increase the affinity for an Fey receptor by modifying one or more amino acids at the following positions: 234, 235, 236, 238, 239, 240, 241 , 243, 244, 245, 247, 248, 249, 252, 254, 255, 256, 258, 262, 263, 264, 265, 267, 268, 269, 270, 272, 276, 278, 280, 283, 285, 286, 289, 290, 292, 293, 294, 295, 296, 298, 299, 301, 303, 305, 307, 309, 312, 313, 315, 320, 322, 324, 325, 326, 327, 329, 330, 331, 332, 333, 334, 335, 337, 338, 340, 360, 373, 376, 378, 382, 388, 389, 398, 414, 416, 419, 430, 43
  • ADCC
  • Exemplary substitutions include 236A, 239D, 239E, 268D, 267E, 268E, 268F, 324T, 332D, and 332E.
  • Exemplary variants include 239D/332E, 236A/332E, 236A/239D/332E, 268F/324T, 267E/268F, 267E/324T, and 267E/268F7324T.
  • Fc modifications that increase binding to an Fey receptor include amino acid modifications at any one or more of amino acid positions 238, 239, 248, 249, 252, 254, 255, 256, 258, 265, 267, 268, 269, 270, 272, 279, 280, 283, 285, 298, 289, 290, 292, 293, 294, 295, 296, 298, 301, 303, 305, 307, 312, 315, 324, 327, 329, 330, 335, 337, 3338, 340, 360, 373, 376, 379, 382, 388, 389, 398, 414, 416, 419, 430, 434, 435, 437, 438 or 439 of the Fc region, wherein the numbering of the residues in the Fc region is that of the EU index as in abat (WO00/42072).
  • Fc modifications that can be made to Fes are those for reducing or ablating binding to FcyR and/or complement proteins, thereby reducing or ablating Fc-mediated effector functions such as ADCC, ADCP, and CDC.
  • Exemplary modifications include but are not limited substitutions, insertions, and deletions at positions 234, 235, 236, 237, 267, 269, 325, and 328, wherein numbering is according to the EU index.
  • Exemplary substitutions include but are not limited to 234G, 235G, 236R, 237K, 267R, 269R, 325L, and 328R, wherein numbering is according to the EU index.
  • An Fc variant may comprise 236R/328R.
  • the Fc region may comprise a non-naturally occurring amino acid residue at additional and/or alternative positions known to one skilled in the art (see, e.g., U.S. Pat. Nos. 5,624,821; 6,277,375; 6,737,056; 6,194,551; 7,317,091; 8,101,720; WO00/42072; WO01/58957; WO02/06919; WO04/016750; WO04/029207; WO04/035752; WO04/074455; WO04/099249; WO04/063351; WO05/070963; WO05/040217, WO05/092925 and WO06/020114).
  • Fc variants that enhance affinity for an inhibitory receptor FcyRllb may also be used.
  • Such variants may provide an Fc fusion protein with immune-modulatory activities related to FcyRllb cells, including for example B cells and monocytes.
  • the Fc variants provide selectively enhanced affinity to FcyRllb relative to one or more activating receptors.
  • Modifications for altering binding to FcyRllb include one or more modifications at a position selected from the group consisting of 234, 235, 236, 237, 239, 266, 267, 268, 325, 326, 327, 328, and 332, according to the EU index.
  • Exemplary substitutions for enhancing FcyRllb affinity include but are not limited to 234D, 234E, 234F, 234W, 235D, 235F, 235R, 235Y, 236D, 236N, 237D, 237N, 239D, 239E, 266M, 267D, 267E, 268D, 268E, 327D, 327E, 328F, 328W, 328Y, and 332E.
  • Exemplary substitutions include 235Y, 236D, 239D, 266M, 267E, 268D, 268E, 328F, 328W, and 328Y.
  • Fc variants for enhancing binding to FcyRllb include 235Y/267E, 236D/267E, 239D/268D, 239D/267E, 267E/268D, 267E/268E, and 267E/328F.
  • the affinities and binding properties of an Fc region for its ligand may be determined by a variety of in vitro assay methods (biochemical or immunological based assays) known in the art including but not limited to, equilibrium methods ⁇ e.g., enzyme-linked immune- absorbent assay (ELISA), or radioimmunoassay (RIA)), or kinetics ⁇ e.g., BIACORE analysis), and other methods such as indirect binding assays, competitive inhibition assays, fluorescence resonance energy transfer (FRET), gel electrophoresis and chromatography ⁇ e.g., gel filtration).
  • in vitro assay methods biochemical or immunological based assays
  • equilibrium methods ⁇ e.g., enzyme-linked immune- absorbent assay (ELISA), or radioimmunoassay (RIA)
  • kinetics e.g., BIACORE analysis
  • indirect binding assays e.g., competitive inhibition assays, fluorescence resonance energy transfer (FRET), gel electro
  • these and other methods may utilize a label on one or more of the components being examined and/or employ a variety of detection methods including but not limited to chromogenic, fluorescent, luminescent, or isotopic labels.
  • detection methods including but not limited to chromogenic, fluorescent, luminescent, or isotopic labels.
  • a detailed description of binding affinities and kinetics can be found in Paul, W. E., ed., Fundamental immunology, 4th Ed., Lippincott-Raven, Philadelphia (1999), which focuses on antibody-immunogen interactions.
  • the antibody is modified to increase its biological half-life.
  • Various approaches are possible. For example, this may be done by increasing the binding affinity of the Fc region for FcRn.
  • one or more of following residues can be mutated: 252, 254, 256, 433, 435, 436, as described in U.S. Pat. No. 6,277,375.
  • Specific exemplary substitutions include one or more of the following: T252L, T254S, and/or T256F.
  • the antibody can be altered within the CHI or CL region to contain a salvage receptor binding epitope taken from two loops of a CH2 domain of an Fc region of an IgG, as described in U.S. Patent Nos. 5,869,046 and 6, 121,022 by Presta et al.
  • variants that increase binding to FcRn and/or improve pharmacokinetic properties include substitutions at positions 259, 308, 428, and 434, including for example 2591, 308F, 428L, 428M, 434S, 434H. 434F, 434Y, and 434M.
  • Other variants that increase Fc binding to FcRn include: 250E, 250Q, 428L, 428F, 250Q/428L (Hinton et al. 2004, J. Biol. Chem. 279(8): 6213-6216, Hinton et al.
  • hybrid IgG isotypes with particular biological characteristics may be used.
  • an IgGl/IgG3 hybrid variant may be constructed by substituting IgGl positions in the CH2 and/or CH3 region with the amino acids from IgG3 at positions where the two isotypes differ.
  • hybrid variant IgG antibody may be constructed that comprises one or more substitutions, e.g., 274Q, 276K, 300F, 339T, 356E, 358M, 384S, 392N, 397M, 4221, 435R, and 436F.
  • an IgGl/IgG2 hybrid variant may be constructed by substituting IgG2 positions in the CH2 and/or CH3 region with amino acids from IgGl at positions where the two isotypes differ.
  • a hybrid variant IgG antibody may be constructed chat comprises one or more substitutions, e.g., one or more of the following amino acid substitutions: 233E, 234L, 235L, -236G (referring to an insertion of a glycine at position 236), and 321 h.
  • IgGl variants with strongly enhanced binding to FcyRIIIa have been identified, including variants with S239D/I332E and S239D/I332E/A330L mutations which showed the greatest increase in affinity for FcyRIIIa, a decrease in FcyRIIb binding, and strong cytotoxic activity in cynomolgus monkeys (Lazar et al. , 2006).
  • IgGl mutants containing L235V, F243L, R292P, Y300L and P396L mutations which exhibited enhanced binding to FcyRIIIa and concomitantly enhanced ADCC activity in transgenic mice expressing human FcyRIIIa in models of B cell malignancies and breast cancer have been identified (Stavenhagen et al., 2007; Nordstrom et al., 2011).
  • Other Fc mutants that may be used include: S298A/E333A/L334A, S239D/I332E, S239D/I332E/A330L, L235V/F243L/R292P/Y300L/ P396L, and M428L/N434S.
  • an Fc is chosen that has reduced binding to FcyRs.
  • An exemplary Fc, e.g., IgGl Fc, with reduced FcyR binding comprises the following three amino acid substitutions: L234A, L235E and G237A.
  • an Fc is chosen that has reduced complement fixation.
  • An exemplary Fc e.g., IgGl Fc, with reduced complement fixation has the following two amino acid substitutions: A330S and P331 S.
  • an Fc is chosen that has essentially no effector function, i.e., it has reduced binding to FcyRs and reduced complement fixation.
  • An exemplary Fc, e.g., IgGl Fc, that is effectorless comprises the following five mutations: L234A, L235E, G237A, A330S and P331 S.
  • An antibody provided herein may be further modified to contain additional nonproteinaceous moieties that are known in the art and readily available.
  • the moieties suitable for derivatization of the antibody include but are not limited to water soluble polymers.
  • Non-limiting examples of water soluble polymers include, but are not limited to, polyethylene glycol (PEG), copolymers of ethylene glycol/propylene glycol, carboxymethylcellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone, poly-1,3- dioxolane, poly-l,3,6-trioxane, ethylene/maleic anhydride copolymer, polyaminoacids (either homopolymers or random copolymers), and dextran or poly(n-vinyl pyrrolidone)polyethylene glycol, propropylene glycol homopolymers, prolypropylene oxide/ethylene oxide copolymers, polyoxyethylated polyols ⁇ e.g., glycerol), polyvinyl alcohol, and mixtures thereof.
  • PEG polyethylene glycol
  • copolymers of ethylene glycol/propylene glycol carboxymethylcellulose
  • dextran polyvinyl alcohol
  • Polyethylene glycol propionaldehyde may have advantages in manufacturing due to its stability in water.
  • the polymer may be of any molecular weight, and may be branched or unbranched.
  • the number of polymers attached to the antibody may vary, and if more than one polymer are attached, they can be the same or different molecules. In general, the number and/or type of polymers used for derivatization can be determined based on considerations including, but not limited to, the particular properties or functions of the antibody to be improved, whether the antibody derivative will be used in a therapy under defined conditions, etc.
  • conjugates of an antibody and nonproteinaceous moiety that may be selectively heated by exposure to radiation are provided.
  • the nonproteinaceous moiety is a carbon nanotube (Kam et al., Proc. Natl. Acad. Sci. USA 102: 11600-11605 (2005)).
  • the radiation may be of any wavelength, and includes, but is not limited to, wavelengths that do not harm ordinary cells, but which heat the nonproteinaceous moiety to a temperature at which cells proximal to the antibody-nonproteinaceous moiety are killed.
  • An antibody can be pegylated to, for example, increase the biological ⁇ e.g., serum) half-life of the antibody.
  • the antibody, or fragment thereof typically is reacted with polyethylene glycol (PEG), such as a reactive ester or aldehyde derivative of PEG, under conditions in which one or more PEG groups become attached to the antibody or antibody fragment.
  • PEG polyethylene glycol
  • the pegylation is carried out via an acylation reaction or an alkylation reaction with a reactive PEG molecule (or an analogous reactive water-soluble polymer).
  • polyethylene glycol is intended to encompass any of the forms of PEG that have been used to derivatize other proteins, such as mono (CI -CIO) alkoxy- or aryloxy-polyethylene glycol or polyethylene glycol-maleimide.
  • the antibody to be pegylated is an aglycosylated antibody. Methods for pegylating proteins are known in the art and can be applied to the antibodies described herein. See for example, EP 0 154 316 by Nishimura et al. and EP0401384 by Ishikawa et al.
  • the present invention also encompasses a human monoclonal antibody described herein conjugated to a therapeutic agent, a polymer, a detectable label or enzyme.
  • the therapeutic agent is a cytotoxic agent.
  • the polymer is polyethylene glycol (PEG).
  • Antibodies may be produced using recombinant methods and compositions, e.g., as described in U.S. Pat. No. 4,816,567.
  • isolated nucleic acid encoding an anti-hemagglutinin antibody described herein is provided.
  • Such nucleic acid may encode an amino acid sequence comprising the VL and/or an amino acid sequence comprising the VH of the antibody ⁇ e.g., the light and/or heavy chains of the antibody).
  • one or more vectors ⁇ e.g., expression vectors) comprising such nucleic acid are provided.
  • a host cell comprising such nucleic acid is provided.
  • a host cell comprises ⁇ e.g., has been transformed with): (1) a vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and an amino acid sequence comprising the VH of the antibody, or (2) a first vector comprising a nucleic acid that encodes an amino acid sequence comprising the VL of the antibody and a second vector comprising a nucleic acid that encodes an amino acid sequence comprising the VH of the antibody.
  • the host cell is eukaryotic, e.g. a Chinese Hamster Ovary (CHO) cell or lymphoid cell ⁇ e.g., Y0, NSO, Sp20 cell).
  • a method of making an anti-hemagglutinin antibody comprises culturing a host cell comprising a nucleic acid encoding the antibody, as provided above, under conditions suitable for expression of the antibody, and optionally recovering the antibody from the host cell (or host cell culture medium).
  • nucleic acid encoding an antibody is isolated and inserted into one or more vectors for further cloning and/or expression in a host cell.
  • nucleic acid may be readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the antibody).
  • Suitable host cells for cloning or expression of antibody-encoding vectors include prokaryotic or eukaryotic cells described herein.
  • antibodies may be produced in bacteria, in particular when glycosylation and Fc effector function are not needed.
  • For expression of antibody fragments and polypeptides in bacteria see, e.g., U.S. Pat. Nos. 5,648,237, 5,789, 199, and 5,840,523. (See also Charlton, Methods in Molecular Biology, Vol. 248 (B.K.C. Lo, ed., Humana Press, Totowa, N.J., 2003), pp. 245-254, describing expression of antibody fragments in E. coli.)
  • the antibody may be isolated from the bacterial cell paste in a soluble fraction and can be further purified.
  • eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for antibody-encoding vectors, including fungi and yeast strains whose glycosylation pathways have been "humanized,” resulting in the production of an antibody with a partially or fully human glycosylation pattern. See Gerngross, Nat. Biotech. 22: 1409-1414 (2004), and Li et al, Nat. Biotech. 24:210-215 (2006).
  • Suitable host cells for the expression of glycosylated antibody are also derived from multicellular organisms (invertebrates and vertebrates). Examples of invertebrate cells include plant and insect cells. Numerous baculoviral strains have been identified which may be used in conjunction with insect cells, particularly for transfection of Spodoptera frugiperda cells.
  • Plant cell cultures can also be utilized as hosts. See, e.g., U.S. Pat. Nos. 5,959,177, 6,040,498, 6,420,548, 7,125,978, and 6,417,429 (describing PLANTIBODIES technology for producing antibodies in transgenic plants).
  • Vertebrate cells may also be used as hosts.
  • mammalian cell lines that are adapted to grow in suspension may be useful.
  • Other examples of useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7); human embryonic kidney line (293 or 293 cells as described, e.g., in Graham et al., J. Gen Virol. 36:59 (1977)); baby hamster kidney cells (BHK); mouse Sertoli cells (TM4 cells as described, e.g., in Mather, Biol. Reprod.
  • monkey kidney cells (CV1); African green monkey kidney cells (VERO-76); human cervical carcinoma cells (HELA); canine kidney cells (MDCK; buffalo rat liver cells (BRL 3A); human lung cells (W138); human liver cells (Hep G2); mouse mammary tumor (MMT 060562); TRI cells, as described, e.g., in Mather et al, Annals N.Y. Acad. Sci. 383 :44-68 (1982); MRC 5 cells; and FS4 cells.
  • Other useful mammalian host cell lines include Chinese hamster ovary (CHO) cells, including DHFR " CHO cells (Urlaub et al, Proc. Natl. Acad. Sci.
  • the antibodies of this invention represent an excellent way for the development of antiviral therapies either alone or in antibody cocktails with additional anti-IAV antibodies for the treatment of human influenza infections in humans.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising the antibodies of the present invention described herein formulated together with a pharmaceutically acceptable carrier.
  • the composition may optionally contain one or more additional pharmaceutically active ingredients, such as another antibody or a therapeutic agent.
  • the pharmaceutical compositions of the invention also can be administered in a combination therapy with, for example, another immune-stimulatory agent, an antiviral agent, or a vaccine, etc..
  • a composition comprises an anti-HA antibody of this invention at a concentration of at least 1 mg/ml, 5 mg/ml, 10 mg/ml, 50 mg/ml, 100 mg/ml, 150 mg/ml, 200 mg/ml, 1-300 mg/ml, or 100-300 mg/ml.
  • the pharmaceutical composition can comprise any number of excipients.
  • Excipients that can be used include carriers, surface active agents, thickening or emulsifying agents, solid binders, dispersion or suspension aids, solubilizers, colorants, flavoring agents, coatings, disintegrating agents, lubricants, sweeteners, preservatives, isotonic agents, and combinations thereof.
  • the selection and use of suitable excipients is taught in Gennaro, ed., Remington: The Science and Practice of Pharmacy, 20th Ed. (Lippincott Williams & Wilkins 2003), the disclosure of which is incorporated herein by reference.
  • a pharmaceutical composition is suitable for intravenous, intramuscular, subcutaneous, parenteral, spinal or epidermal administration ⁇ e.g., by injection or infusion).
  • the active compound can be coated in a material to protect it from the action of acids and other natural conditions that may inactivate it.
  • parenteral administration means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion.
  • an antibody of the present invention described herein can be administered via a non-parenteral route, such as a topical, epidermal or mucosal route of administration, e.g., intranasally, orally, vaginally, rectally, sublingually or topically.
  • a non-parenteral route such as a topical, epidermal or mucosal route of administration, e.g., intranasally, orally, vaginally, rectally, sublingually or topically.
  • the pharmaceutical composition of the invention can be in the form of pharmaceutically acceptable salts.
  • a "pharmaceutically acceptable salt” refers to a salt that retains the desired biological activity of the parent compound and does not impart any undesired toxicological effects. Examples of such salts include acid addition salts and base addition salts. Acid addition salts include those derived from nontoxic inorganic acids, such as hydrochloric, nitric, phosphoric, sulfuric, hydrobromic, hydroiodic, phosphorous and the like, as well as from nontoxic organic acids such as aliphatic mono- and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxy alkanoic acids, aromatic acids, aliphatic and aromatic sulfonic acids and the like.
  • Base addition salts include those derived from alkaline earth metals, such as sodium, potassium, magnesium, calcium and the like, as well as from nontoxic organic amines, such as ⁇ , ⁇ '-dibenzylethylenediamine, N-methylglucamine, chloroprocaine, choline, diethanolamine, ethylenediamine, procaine and the like.
  • the pharmaceutical composition of the present invention can be in the form of sterile aqueous solutions or dispersions. It can also be formulated in a microemulsion, liposome, or other ordered structure suitable to high drug concentration.
  • An antibody of the present invention described herein can be administered as a sustained release formulation, in which case less frequent administration is required. Dosage and frequency vary depending on the half-life of the antibody in the patient. In general, human antibodies show the longest half-life, followed by humanized antibodies, chimeric antibodies, and nonhuman antibodies. The dosage and frequency of administration can vary depending on whether the treatment is prophylactic or therapeutic. In prophylactic applications, a relatively low dosage is administered at relatively infrequent intervals over a long period of time. Some patients continue to receive treatment for the rest of their lives. In therapeutic applications, a relatively high dosage at relatively short intervals is sometimes required until progression of the disease is reduced or terminated, and preferably until the patient shows partial or complete amelioration of symptoms of disease. Thereafter, the patient can be administered a prophylactic regime.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the subject being treated and the particular mode of administration and will generally be that amount of the composition which produces a therapeutic effect. Generally, out of one hundred percent, this amount will range from about 0.01% to about 99% of active ingredient, preferably from about 0.1% to about 70%, most preferably from about 1% to about 30% of active ingredient in combination with a pharmaceutically acceptable carrier.
  • Dosage regimens can be adjusted to provide the optimum desired response (e.g., a therapeutic response). For example, a single bolus can be administered, several divided doses can be administered over time or the dose can be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage.
  • Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • antibody can be administered as a sustained release formulation, in which case less frequent administration is required.
  • the dosage ranges from about 0.0001 to 100 mg/kg, and more usually 0.01 to 5 mg/kg, of the host body weight.
  • dosages can be 0.3 mg/kg body weight, 1 mg/kg body weight, 3 mg/kg body weight, 5 mg/kg body weight or 10 mg/kg body weight or within the range of 1-10 mg/kg.
  • An exemplary treatment regime entails administration once per week, once every two weeks, once every three weeks, once every four weeks, once a month, once every 3 months or once every three to 6 months.
  • Preferred dosage regimens for an anti-HA antibody of the invention include 1 mg/kg body weight or 3 mg/kg body weight via intravenous administration, with the antibody being given using one of the following dosing schedules: (i) every four weeks for six dosages, then every three months; (ii) every three weeks; (iii) 3 mg/kg body weight once followed by 1 mg/kg body weight every three weeks.
  • dosage is adjusted to achieve a plasma antibody concentration of about 1-1000 ⁇ g /ml and in some methods about 25-300 ⁇ g /ml.
  • a “therapeutically effective dosage” of an anti-HA antibody of the invention preferably results in a decrease in severity of disease symptoms, an increase in frequency and duration of disease symptom-free periods, or a prevention of impairment or disability due to the disease affliction.
  • a "therapeutically effective dosage” preferably inhibits IAV virus replication or uptake by host cells by at least about 20%), more preferably by at least about 40%, even more preferably by at least about 60%), and still more preferably by at least about 80%> relative to untreated subj ects.
  • a therapeutically effective amount of a therapeutic compound can neutralize IAV virus, or otherwise ameliorate symptoms in a subject, which is typically a human or can be another mammal.
  • the pharmaceutical composition can be a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. See, e.g., Sustained and Controlled Release Drug Delivery Systems, J. R. Robinson, ed., Marcel Dekker, Inc., New York, 1978.
  • Therapeutic compositions can be administered via medical devices such as (1) needleless hypodermic injection devices (e.g., US 5,399, 163; 5,383,851; 5,312,335;
  • medical devices such as (1) needleless hypodermic injection devices (e.g., US 5,399, 163; 5,383,851; 5,312,335;
  • transdermal devices (US 4,486, 194); (4) infusion apparati (US 4,447,233 and 4,447,224); and (5) osmotic devices (US 4,439,196 and 4,475, 196); the disclosures of which are incorporated herein by reference.
  • the human monoclonal antibodies of the invention described herein can be formulated to ensure proper distribution in vivo.
  • the therapeutic compounds of the invention can be formulated in liposomes, which may additionally comprise targeting moieties to enhance selective transport to specific cells or organs. See, e.g. US 4,522,811; 5,374,548; 5,416,016; and 5,399,331; V.V. Ranade (1989) /. Clin. Pharmacol. 29:685; Umezawa et al, (1988)
  • the current anti-viral treatments e.g. oseltamivir/Tamiflu, amantadine/rimantadine
  • the current anti-viral treatments are sub-optimal with increasing incidence of resistance and a limited therapeutic window (must start ⁇ 48 hours after symptom onset)
  • a limited therapeutic window must start ⁇ 48 hours after symptom onset
  • Monoclonal antibodies continue to be a growing class of drugs in-part due to their high degree of specificity, limited off-target effects, and superb safety profile.
  • the antibodies, compositions and formulations described herein can be used to neutralize influenza virus and thereby treating influenza infections.
  • the anti-HA antibodies described herein can be used to neutralize IAV virus.
  • the neutralizing of the IAV virus can be done via (i) inhibiting IAV virus binding to a target cell; (ii) inhibiting IAV virus uptake by a target cell; (iii) inhibiting IAV virus replication; and (iv) inhibiting IAV virus particles release from infected cells.
  • One skilled in the art possesses the ability to perform any assay to assess neutralization of IAV virus.
  • the neutralizing properties of antibodies may be assessed by a variety of tests, which all may assess the consequences of (i) inhibition of IAV virus binding to a target cell; (ii) inhibition of IAV virus uptake by a target cell; (iii) inhibition of IAV virus replication; and (iv) inhibition of IAV virus particles release from infected cells.
  • implementing different tests may lead to the observation of the same consequence, i.e. the loss of infectivity of the IAV virus.
  • the present invention provides a method of neutralizing IAV virus in a subject comprising administering to the subject a therapeutically effect amount of the antibody of the present invention described herein.
  • Another aspect of the present invention provides a method of treating an IAV-related disease.
  • Such method includes therapeutic (following IAV infection) and prophylactic (prior to IAV exposure, infection or pathology).
  • therapeutic and prophylactic methods of treating an individual for an IAV infection include treatment of an individual having or at risk of having an IAV infection or pathology, treating an individual with an IAV infection, and methods of protecting an individual from an IAV infection, to decrease or reduce the probability of an IAV infection in an individual, to decrease or reduce susceptibility of an individual to an IAV infection, or to inhibit or prevent an IAV infection in an individual, and to decrease, reduce, inhibit or suppress transmission of an IAV from an infected individual to an uninfected individual.
  • Such methods include administering an antibody of the present invention or a composition comprising the antibody disclosed herein to therapeutically or prophylactically treat (vaccinate or immunize) an individual having or at risk of having an IAV infection or pathology. Accordingly, methods can treat the IAV infection or pathology, or provide the individual with protection from infection ⁇ e.g., prophylactic protection).
  • a method of treating an IAV-related disease comprises administering to an individual in need thereof an anti-HA antibody or therapeutic composition disclosed herein in an amount sufficient to reduce one or more physiological conditions or symptom associated with an IAV infection or pathology, thereby treating the IAV-related disease.
  • an anti-HA antibody or therapeutic composition disclosed herein is used to treat an IAV-related disease.
  • Use of an anti-HA antibody or therapeutic composition disclosed herein treats an IAV-related disease by reducing one or more physiological conditions or symptom associated with an IAV infection or pathology.
  • administration of an anti-HA or therapeutic composition disclosed herein is in an amount sufficient to reduce one or more physiological conditions or symptom associated with an IAV infection or pathology, thereby treating the IAV-based disease.
  • administration of an anti-HA antibody or therapeutic composition disclosed herein is in an amount sufficient to increase, induce, enhance, augment, promote or stimulate IAV clearance or removal; or decrease, reduce, inhibit, suppress, prevent, control, or limit transmission of IAV to another individual.
  • One or more physiological conditions or symptom associated with an IAV infection or pathology will respond to a method of treatment disclosed herein.
  • the symptoms of IAV infection or pathology vary, depending on the phase of infection.
  • the anti-HA antibody described herein can be used in various detection methods, for use in, e.g., monitoring the progression of an IAV infection; monitoring patient response to treatment for such an infection, etc.
  • the present disclosure provides methods of detecting an HA polypeptide in a biological sample obtained from an individual. The methods generally involve: a) contacting the biological sample with a subject anti-HA antibody; and b) detecting binding, if any, of the antibody to an epitope present in the sample.
  • the antibody comprises a detectable label.
  • the level of HA polypeptide detected in the biological sample can provide an indication of the stage, degree, or severity of an IAV infection.
  • the level of HA polypeptide detected in the biological sample can provide an indication of the individual's response to treatment for an IAV infection.
  • the antibodies described herein can be used together with one or more of other anti- influenza virus antibodies to neutralize influenza virus and thereby treating influenza infections.
  • hmAbs such as 1F1 (Tsibane T, et al. 2012. PLoS Pathog 8:el003067)and CH65 (Whittle JR, et al. 2011. Proc Natl Acad Sci U S A 108: 14216-14221), which bind multiple HI isolates; hmAbs such as F10 (Sui J, et al Hwang et al. 2009. Nat Struct Mol Biol 16:265- 273) and CR6261 (Ekiert DC, et al. 2009.
  • antibody as referred to herein includes whole antibodies and any antigen binding fragment or single chains thereof.
  • Whole antibodies are glycoproteins comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds.
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region.
  • the heavy chain constant region is comprised of three domains, CH I , CH2 and CH3.
  • Each light chain is comprised of a light chain variable region (abbreviated herein as V L ) and a light chain constant region.
  • the light chain constant region is comprised of one domain, .
  • VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR).
  • CDR complementarity determining regions
  • FR framework regions
  • Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the heavy chain variable region CDRs and FRs are HFR1, HCDR1, HFR2, HCDR2, HFR3, HCDR3, HFR4.
  • the light chain variable region CDRs and FRs are LFR1, LCDR1, LFR2, LCDR2, LFR3, LCDR3, LFR4.
  • variable regions of the heavy and light chains contain a binding domain that interacts with an antigen.
  • the constant regions of the antibodies can mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system ⁇ e.g., effector cells) and the first component (Clq) of the classical complement system.
  • antigen-binding fragment or portion of an antibody refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen ⁇ e.g., an HA of influenza A virus). It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody.
  • binding fragments encompassed within the term "antigen-binding fragment or portion" of an antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CHI domains; (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fab' fragment, which is essentially an Fab with part of the hinge region (see, FUNDAMENTAL FMMUNOLOGY (Paul ed., 3 rd ed.
  • the two domains of the Fv fragment, VL and VH are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883).
  • single chain Fv single chain Fv
  • Such single chain antibodies are also intended to be encompassed within the term "antigen-binding fragment or portion" of an antibody.
  • an "isolated antibody”, as used herein, is intended to refer to an antibody that is substantially free of other antibodies having different antigenic specificities ⁇ e.g., an isolated antibody that specifically binds to an HA of influenza A virus is substantially free of antibodies that specifically bind antigens other than the HA).
  • An isolated antibody can be substantially free of other cellular material and/or chemicals.
  • monoclonal antibody or “monoclonal antibody composition” as used herein refer to a preparation of antibody molecules of single molecular composition.
  • a monoclonal antibody composition displays a single binding specificity and affinity for a particular epitope.
  • human antibody is intended to include antibodies having variable regions in which both the framework and CDR regions are derived from human germline immunoglobulin sequences. Furthermore, if the antibody contains a constant region, the constant region also is derived from human germline immunoglobulin sequences.
  • the human antibodies of the invention can include amino acid residues not encoded by human germline immunoglobulin sequences ⁇ e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo).
  • the term "human antibody”, as used herein is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
  • human monoclonal antibody refers to antibodies displaying a single binding specificity, which have variable regions in which both the framework and CDR regions are derived from human germline immunoglobulin sequences.
  • the human monoclonal antibodies can be produced by a hybridoma which includes a B cell obtained from a transgenic nonhuman animal, e.g., a transgenic mouse, having a genome comprising a human heavy chain transgene and a light chain transgene fused to an immortalized cell.
  • recombinant human antibody includes all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as (a) antibodies isolated from an animal (e.g., a mouse) that is transgenic or transchromosomal for human immunoglobulin genes or a hybridoma prepared therefrom (described further below), (b) antibodies isolated from a host cell transformed to express the human antibody, e.g., from a transfectoma, (c) antibodies isolated from a recombinant, combinatorial human antibody library, and (d) antibodies prepared, expressed, created or isolated by any other means that involve splicing of human immunoglobulin gene sequences to other DNA sequences.
  • Such recombinant human antibodies have variable regions in which the framework and CDR regions are derived from human germline immunoglobulin sequences.
  • such recombinant human antibodies can be subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo.
  • isotype refers to the antibody class ⁇ e.g., IgM or IgGl) that is encoded by the heavy chain constant region genes.
  • the phrases "an antibody recognizing an antigen” and “an antibody specific for an antigen” are used interchangeably herein with the term “an antibody which binds specifically to an antigen.”
  • human antibody derivatives refers to any modified form of the human antibody, e.g., a conjugate of the antibody and another agent or antibody.
  • humanized antibody is intended to refer to antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences. Additional framework region modifications can be made within the human framework sequences.
  • chimeric antibody is intended to refer to antibodies in which the variable region sequences are derived from one species and the constant region sequences are derived from another species, such as an antibody in which the variable region sequences are derived from a mouse antibody and the constant region sequences are derived from a human antibody.
  • the term can also refer to an antibody in which its variable region sequence or CDR(s) is derived from one source (e.g., an IgAl antibody) and the constant region sequence or Fc is derived from a different source (e.g., a different antibody, such as an IgG, IgA2, IgD, IgE or IgM antibody).
  • affinity refers to the strength of the sum total of noncovalent interactions between a single binding site of a molecule (e.g., an antibody) and its binding partner (e.g., an antigen).
  • binding affinity refers to intrinsic binding affinity which reflects a 1 : 1 interaction between members of a binding pair (e.g., antibody and antigen).
  • the affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (KD). Affinity can be measured by common methods known in the art, including those described herein.
  • an antibody that "specifically binds to an HA of influenza A virus” refers to an antibody that binds to an HA of influenza A virus but does not substantially bind to non-influenza A virus HA.
  • an antibody that "specifically binds to an HA of influenza A virus HI subtype" refers to an antibody that binds to an HA of influenza A virus HI subtype but does not substantially bind to other subtype of influenza A virus HA.
  • the antibody binds to the HA with "high affinity", namely with a KD of 1 X 10 "7 M or less, more preferably 5 x 10 "8 M or less, more preferably 3 x 10 "8 M or less, more preferably 1 x 10 "8 M or less, more preferably 5 x 10 "9 M or less or even more preferably 1 x 10 "9 M or less.
  • the term "does not substantially bind" to a protein or cells means does not bind or does not bind with a high affinity to the protein or cells, i.e.
  • K asS oc or "K a ", as used herein, is intended to refer to the association rate of a particular antibody-antigen interaction
  • K d i S or "K d ,” as used herein, is intended to refer to the dissociation rate of a particular antibody- antigen interaction
  • KD is intended to refer to the dissociation constant, which is obtained from the ratio of Kd to K a (i.e., Kd/K a ) and is expressed as a molar concentration (M).
  • KD values for antibodies can be determined using methods well established in the art. A preferred method for determining the KD of an antibody is by using surface plasmon resonance, preferably using a biosensor system such as a Biacore ® system.
  • epitope refers to an antigenic determinant that interacts with a specific antigen binding site in the variable region of an antibody molecule known as a paratope.
  • a single antigen may have more than one epitope. Thus, different antibodies may bind to different areas on an antigen and may have different biological effects.
  • epitope also refers to a site on an antigen to which B and/or T cells respond. It also refers to a region of an antigen that is bound by an antibody.
  • Epitopes may be defined as structural or functional. Functional epitopes are generally a subset of the structural epitopes and have those residues that directly contribute to the affinity of the interaction.
  • Epitopes may also be conformational, that is, composed of non-linear amino acids.
  • epitopes may include determinants that are chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl groups, or sulfonyl groups, and, in certain embodiments, may have specific three-dimensional structural characteristics, and/or specific charge characteristics.
  • An epitope typically includes at least 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14 or 15 amino acids in a unique spatial conformation.
  • epitope mapping Methods for determining what epitopes are bound by a given antibody (i.e., epitope mapping) are well known in the art and include, for example, immunoblotting and immune-precipitation assays, wherein overlapping or contiguous peptides from an HA protein are tested for reactivity with a given antibody.
  • Methods of determining spatial conformation of epitopes include techniques in the art and those described herein, for example, x-ray crystallography and 2-dimensional nuclear magnetic resonance (see, e.g. , Epitope Mapping Protocols in Methods in Molecular Biology, Vol. 66, G. E. Morris, Ed. (1996)).
  • epitopope mapping refers to the process of identification of the molecular determinants for antibody-antigen recognition.
  • binds to an epitope or “recognizes an epitope” with reference to an antibody or antibody fragment refers to continuous or discontinuous segments of amino acids within an antigen. Those of skill in the art understand that the terms do not necessarily mean that the antibody or antibody fragment is in direct contact with every amino acid within an epitope sequence.
  • the term "binds to the same epitope" with reference to two or more antibodies means that the antibodies bind to the same, overlapping or encompassing continuous or discontinuous segments of amino acids.
  • Those of skill in the art understand that the phrase "binds to the same epitope” does not necessarily mean that the antibodies bind to or contact exactly the same amino acids.
  • the precise amino acids which the antibodies contact can differ.
  • a first antibody can bind to a segment of amino acids that is completely encompassed by the segment of amino acids bound by a second antibody.
  • a first antibody binds one or more segments of amino acids that significantly overlap the one or more segments bound by the second antibody.
  • such antibodies are considered to "bind to the same epitope.”
  • Antibodies that "compete with another antibody for binding to a target” refer to antibodies that inhibit (partially or completely) the binding of the other antibody to the target. Whether two antibodies compete with each other for binding to a target, i.e., whether and to what extent one antibody inhibits the binding of the other antibody to a target, may be determined using known competition experiments. In certain embodiments, an antibody competes with, and inhibits binding of another antibody to a target by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or 100%. The level of inhibition or competition may be different depending on which antibody is the "blocking antibody” ⁇ i.e., the cold antibody that is incubated first with the target).
  • Competing antibodies bind to the same epitope, an overlapping epitope or to adjacent epitopes ⁇ e.g., as evidenced by steric hindrance).
  • an immune response refers to a biological response within a vertebrate against foreign agents, which response protects the organism against these agents and diseases caused by them.
  • An immune response is mediated by the action of a cell of the immune system (for example, a T lymphocyte, B lymphocyte, natural killer (NK) cell, macrophage, eosinophil, mast cell, dendritic cell or neutrophil) and soluble macromolecules produced by any of these cells or the liver (including antibodies, cytokines, and complement) that results in selective targeting, binding to, damage to, destruction of, and/or elimination from the vertebrate's body of invading pathogens, cells or tissues infected with pathogens, cancerous or other abnormal cells, or, in cases of autoimmunity or pathological inflammation, normal human cells or tissues.
  • An immune reaction includes, e.g., activation or inhibition of a T cell, e.g., an effector T cell or a Th cell, such as a CD4+ or CD
  • detectable label refers to a molecule capable of detection, including, but not limited to, radioactive isotopes, fluorescers, chemiluminescers, chromophores, enzymes, enzyme substrates, enzyme cofactors, enzyme inhibitors, chromophores, dyes, metal ions, metal sols, ligands (e.g., biotin, avidin, streptavidin or haptens), intercalating dyes and the like.
  • fluorescer refers to a substance or a portion thereof which is capable of exhibiting fluorescence in the detectable range.
  • the term "subject" refers to an animal.
  • the animal is a mammal.
  • a subject also refers to for example, primates (e.g., humans), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds and the like.
  • the subject is a human.
  • the term "therapeutically effective amount" of a compound of the present invention refers to an amount of the compound of the present invention that will elicit the biological or medical response of a subject, or ameliorate symptoms, slow or delay disease progression, or prevent a disease, etc. In one embodiment, the term refers to the amount that inhibits or reduces microbial colonization or infection. In one embodiment, the term refers to the amount that inhibits or reduces infection, or prevent or destroying the formation of bacterial biofilms. When applied to an individual active ingredient, administered alone, the term refers to that ingredient alone. When applied to a combination, the term refers to combined amounts of the active ingredients that result in the therapeutic effect, whether administered in combination, serially or simultaneously.
  • the term "pharmaceutically acceptable carrier or excipient” refers to a carrier medium or an excipient which does not interfere with the effectiveness of the biological activity of the active ingredient(s) of the composition and which is not excessively toxic to the host at the concentrations at which it is administered.
  • a pharmaceutically acceptable carrier or excipient is preferably suitable for topical formulation.
  • the term includes, but is not limited to, a solvent, a stabilizer, a solubilizer, a tonicity enhancing agent, a structure-forming agent, a suspending agent, a dispersing agent, a chelating agent, an emulsifying agent, an anti-foaming agent, an ointment base, an emollient, a skin protecting agent, a gel-forming agent, a thickening agent, a pH adjusting agent, a preservative, a penetration enhancer, a complexing agent, a lubricant, a demulcent, a viscosity enhancer, a bioadhesive polymer, or a combination thereof.
  • treating refers in one embodiment, to ameliorating the disease or disorder (i.e., arresting or reducing the development of the disease or at least one of the clinical symptoms thereof). In another embodiment “treating” or “treatment” refers to ameliorating at least one physical parameter, which may not be discernible by the patient. In yet another embodiment, “treating” or “treatment” refers to modulating the disease or disorder, either physically, (e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter), or both. In yet another embodiment, “treating” or “treatment” refers to preventing or delaying the onset or development or progression of the disease or disorder.
  • Canine Madin-Darby canine kidney (MDCK; ATCC CCL-34) and human embryonic kidney (HEK293T; ATCC CRL-11268) cells were grown at 37 °C with 5% C0 2 in Dulbecco's modified Eagle's medium (DMEM; Mediatech, Inc.), 10% fetal bovine serum (FBS), and 1% PSG (penicillin, 100 units/ml; streptomycin 100 ⁇ g/ml; L-glutamine, 2 mM) (Nogales A, et al. 2014. J Virol 88: 10525-10540).
  • DMEM Dulbecco's modified Eagle's medium
  • FBS fetal bovine serum
  • PSG penicillin, 100 units/ml
  • L-glutamine 2 mM
  • influenza A and B viruses were propagated in MDCK cells: A/California/04/2009 (pHlNl) wild-type (WT) and mCherry-expressing viruses (Nogales A, et al. 2015. Virus Res 213 :69-81; and Nogales A, et al. 2014. Virology 476C:206-216); A/Puerto Rico/08/1934 HlNl (PR8 HlNl) WT and mCherry-expressing viruses (Nogales A, et al. 2014.
  • Virology 476C:206-216 A/Texas/36/91 HlNl (TX HlNl), A/New Caledonia/20/99 HlNl (NC HlNl), A/Wyoming/3/2003 H3N2 (H3N2) WT and mCherry-expressing viruses; and B/Brisbane/60/2008 (IBV) WT and mCherry-expressing viruses (Nogales A, et al. 2015. Virus Res 213 :69-81).
  • virus stocks were diluted in phosphate buffered saline (PBS), 0.3% bovine albumin (BA) and 1% PS (PBS/BA/PS).
  • Peripheral blood was obtained from a healthy subject prior to, seven days, and one month after receiving the 2014-2015 seasonal inactivated quadrivalent influenza vaccine (A/California/07/2009 (HlNl) pdm09-like virus, A/Texas/50/2012 (H3N2)-like virus, B/Massachusetts/2/2012-like virus, B/Brisbane/60/2008-like virus) as standard-of-care at the University of Rochester Medical Center.
  • the subject provided signed written informed consent. All procedures and methods were approved by the Research Subjects Review Board at the University of Rochester Medical Center and all experiments were performed in accordance with relevant guidelines and regulations.
  • PBMC and plasma was isolated using CPT tubes (Becton Dickinson, Franklin Lakes, NJ, USA). Fresh PBMC from seven days after immunization were stained for flow cytometry as previously described with anti-CD45- Qdot800 (HI30, Invitrogen, Carlsbad, CA), anti-CD 19-APC-Cy7 (SJ25C1, BD Biosciences, San Jose, CA), anti-CD20-AlexaFluor 700 (2H7, Biolegend, San Diego, CA), anti-CD3- PacificOrange (UCHT1, Invitrogen), anti-IgD-FITC (IA6-2, BD), anti-CD27-Qdot655 (CLB- 27/1, Invitrogen), anti-CD4-Qdot705 (S3.5, Invitrogen), anti-CD38-Qdot605 (HIT2, Invitrogen), anti-CD126-PE (M5, BD), and Live/Dead fixable aqua dead cell stain (Invitrog
  • Plasmablasts were single cell sorted with a FACSAria (BD Biosciences) directly into 96-well PCR plates (Bio-Rad, Hercules, CA) containing 4 pL/well 0.5X PBS with lO mM DTT (Invitrogen), and 8 U RiboLock (Therm oFisher) RNAse inhibitor. Plates were sealed with MICROSEAL F FILM (Bio-Rad) and immediately frozen at -80 °C until used for RT-PCR. cDNA was synthesized and semi-nested RT-PCR for IgH, 3 ⁇ 4 ⁇ , and IgK V gene transcripts was performed as previously described (Kobie JJ, et al. 2015.
  • Purified PCR products were sequenced at Genewiz Sequences and analyzed by IgBlast (www.ncbi.nlm.nih.gov/igblast) and FMGT/V- QUEST (www.imgt.org/EVIGT_vquest) to identify germline V(D)J gene segments with highest identity and determine sequence properties.
  • Expression vector cloning and transfection of human HEK293T cells ATCC, Manassas, VA were performed as previously described (Kobie JJ, et al. 2015. Monoclon Antib Immunodiagn Immunother 34:65-72; and Tiller T, et al. 2008.
  • IgG was purified from culture supernatant using MAGNA PROTEIN G beads (Promega, Madison, WI). 1069 D6 is a human IgGl mAb that was used as an isotype control.
  • PBMC peripheral blood mononuclear cells
  • the resulting cDNA was used in subsequent PCR using Platinum Taq High Fidelity Polymerase (Invitrogen, Carlsbad, CA) and a touchdown PCR protocol starting with a 95 °C for 5 min of denaturing, then 2 cycles of 96 °C for 30 sec, 62 °C for 30 sec, and 68 °C for 1 min. The annealing temperature was dropped 2 °C for every other cycle until 55 °C which was used for the final 32 cycles. A final extension was performed at 68 °C for 10 min before holding at 12 °C. Degenerate primers were designed based on Sheid et al. 2011, Science 333, 1633-1637 with supplemental primers for VH2, IgM (Richardson, C.
  • PCR products were submitted to the University of Rochester Genomics Research Center where Qubit Fluorometric quantitation (Therm oFisher) and Bioanalyzer (Agilent Technologies, Santa Clara, CA) sizing, quantitation and quality control was performed prior to normalizing to 2 nM and flowcell hybridization and cluster generation for the MiSeq system (Illumina, Inc., San Diego, CA). Paired end reads (300 x 325 bp) were made.
  • ELISA plates (NUNC MAXISORP, Thermo Fisher Scientific, Grand Island, NY) were coated with recombinant HA proteins (Protein Sciences, Meriden, CT) or RSV fusion (F) Protein at 0.5 ⁇ g/mL, hMAbs were diluted in PBS, and binding detected with horseradish peroxidase (HRP)-conjugated anti-human IgG (Jackson ImmunoResearch, West Grove, PA). In select ELISAs increasing concentrations of urea was add to ELISA plate and incubated for 15 min at room temperature prior to detection with anti-IgG-HRP to evaluate avidity.
  • HRP horseradish peroxidase
  • the MPLEX-FLU assay immunoglobulin quantification method was performed as previously described (Wang J, et al. 2015. PLoS One 10:e0129858). Assays were performed in 96 well black-walled microtiter-plates (Millipore, Billerica, MA). Just prior to assay, the coupled beads were vortexed for 15 seconds and diluted to 50 beads of each bead region per ⁇ and added at 25 ⁇ beads per well. All plasma and hmAb dilutions and washes were performed using PBS (pH 7.2) containing 0.1% BSA (MP Biomedical, LLC, France) and 0.1% Brij-35 (Thermo Scientific, Waltham, MA).
  • beads in each well were suspended with 100 ⁇ LUMINEX driving solution (Luminex, Austin, TX) and analyzed on a MAGPLX multiplex reader (Luminex, Austin, TX), and results expressed as median fluorescence intensity (MFI). Binding affinity studies using surface plasmon resonance.
  • KPF1 The binding affinity of KPF1 to recombinant (r)HA of pHlNl A/California/04/2009 (Protein Sciences Corp., Meriden, CT) was determined by SPR experiments performed with a Biacore T200 optical biosensor (Biacore, Uppsala, Sweden) at 25 °C.
  • KPF1 50 nM was flowed across flow cell 2 of a Series S Sensor Chip Protein G (GE HealthCare, Uppsala, Sweden) using 60 sec contact time, 10 ⁇ /min flow rate, and 30 sec for stabilization, capturing approximately 1800 resonance units (RU).
  • Flow cell 1 was left blank (Protein G only) to serve as a reference.
  • pHlNl rHA was used to analyze binding with a 90 s contact time, 75 ⁇ /m flow rate, and 700 sec dissociation time.
  • the sensor surface was regenerated by repeated washes with 10 mM glycine (pH 1.5) at a flow rate of 30 ⁇ /min.
  • Each binding curve was analyzed after correcting for non-specific binding by subtraction of the signals obtained from the negative-control flow channel and buffer injections (Myszka, D. G. Improving biosensor analysis.
  • Virus neutralization assays were performed with WT and mCherry-expressing viruses as previously described (Nogales A, et al. 2015. Virus Res 213 :69-81; and Nogales A, et al. 2014. Virology 476C:206-216). Briefly, KPF1 hmAb or IgGl isotype control hmAb were serially 2-fold diluted in PBS using 96-well plates (starting concentration of 200 ⁇ g). One hundred PFUs of each virus were then added to the hmAb dilutions and incubated for 1 h at room temperature.
  • MDCK cells (96-well plate format, 5 ⁇ 10 4 cells/well, triplicates) were then infected with the hmAb-virus mixture for 1 h at room temperature. After viral adsorption, cells were maintained in p.i. medium, with 1 ⁇ g/ml TPCK-treated trypsin (Martinez-Sobrido, L. & Garcia-Sastre, A. Generation of recombinant influenza virus from plasmid DNA. Journal of visualized experiments: JoVE, https://doi.org/10.3791/2057 (2010)) and incubated at 33 °C. For the fluorescence-based microneutralization assays, at 24-48 h p.i.
  • mice Five to seven-week-old female C57BL/6 mice were purchased from the National Cancer Institute (NCI) and maintained in the animal care facility at University of Rochester under specific pathogen-free conditions. All animal protocols were approved by the University of Rochester Committee of Animal Resources and complied with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Research Council (National Research Council (U.S.). Committee for the Update of the Guide for the Care and Use of Laboratory Animals., Institute for Laboratory Animal Research (U.S.), National Academys Press (U.S.). 2011. Guide for the care and use of laboratory animals, 8th ed. National Academys Press, Washington, D.C).
  • NCI National Cancer Institute
  • mice were anesthetized intraperitoneally (i.p.) with 2,2,2-tribromoethanol (Avertin; 240 mg/kg of body weight) and inoculated intranasally (i.n.) with 10X the mouse lethal dose 50 (MLD50) of pHlNl in a final volume of 30 ⁇ .
  • MLD50 mouse lethal dose 50
  • animals were monitored daily for morbidity (body weight loss) and mortality (survival). Mice showing more that 25% loss of body weight were considered to have reached the experimental endpoint and were humanely euthanized.
  • mice in groups of 11 were weighed and administered i.p.
  • Viral replication (pHlNl, PR8 H1N1, TX H1N1, and NC H1N1) was determined by measuring viral titers in the lungs of infected mice at days 2 and 4 p.i. To that end, three mice from each group were euthanized and lungs were collected and homogenized.
  • Virus titers were determined by immunofocus assay (fluorescent focus-forming units, FFU/ml) (Nogales A, et al. 2014. J Virol 88: 10525-10540 and Nogales A, et al. 2016. J Virol 90:6291-6302) using the anti-NP mAb FIB-65 (ATTC) and a FITC-conjugated anti-mouse secondary Ab (Dako).
  • Geometric mean titers and data representation were performed using (GRAPFIPAD PRISM, v7.0).
  • All the MARMs were selected by incubating the pHlNl or TX H1N1 influenza viruses under increasing concentrations of KPF1 hmAb. Briefly, MDCK cells were infected at low multiplicity of infection (MOI 0.01) with the pHlNl in 24 well-plate. After 1 h of adsorption, p.i. medium containing the KPF1 hmAb was added to the wells. The plates were incubated at 33 °C for 2 to 3 days and observed daily for cytopathic effect (CPE). Once the infected cells exhibited more than 70% of CPE, tissue culture supernatants (TCS) were collected and used to infect fresh MDCK cells (MOI 0.01) as described above.
  • CPE cytopathic effect
  • a polymerase I-driven pPolI plasmid containing the pHlNl HA (Baker, S. F. et al. J Virol 87, 8591-8605, https://doi.org/10.1128/JVI.01081-13 (2013)) was used as template to introduce, using site-directed mutagenesis, the amino acid changes E129K, K180N, K180Q, and E129K/K180N. Then, the Bbsl and Pmll restriction sites were used to subclone the HA fragment containing the amino acid substitutions into a polymerase II-driven pCAGGS protein expression plasmids (Niwa, H., et al.
  • confluent monolayers of MDCK cells were mock-infected or infected (MOI 0.01) with WT or the MARMs viruses.
  • MOI 0.01 the MARMs viruses.
  • cells were fixed with 4% PFA and permeabilized with 0.5% Triton X-100 in PBS for 15 min at room temperature.
  • Cells were then incubated with hmAb KPFl or with the pHlNl HA mouse mAb 29E3 (Manicassamy B, et al. 2010. PLoS Pathog 6:el000745) or against P (HB-65) for 1 hour at 37 °C.
  • the cells were incubated with FITC-conjugated secondary anti-mouse Ab (Dako) and 4',6-diamidino-2-phenylindole (DAPI; Research Organics) for 1 hour at 37 °C. Cells were visualized and photographed using a fluorescence microscope (Olympus 1X81) and camera (Q IMAGING, RETIGA 2000R) with a xlO objective.
  • FITC-conjugated secondary anti-mouse Ab Dako
  • DAPI 4',6-diamidino-2-phenylindole
  • HEK293T cells were transiently transfected, using lipofectamine 2000, with 0.5 ⁇ g of the indicated pCAGGS plasmids and at 24 hours post-transfection, HA expression was analyzed by IF A as indicated above, using the hmAb KPFl or a goat pHlNl anti-HA polyclonal antibody as internal control (BEI Resources NR-15696).
  • Viral RT-PCR Viral RT-PCR
  • RNA from infected MDCK cells was collected at 36 h p.i. and purified using TRIZOL reagent (Invitrogen) according to the manufacturer's specifications.
  • cDNA synthesis for HA and NA mRNAs was performed using Superscript® II Reverse Transcriptase (Invitrogen) and an oligo dT oligonucleotide (Invitrogen). Further, cDNAs were used as templates for PCR with primers specific for the viral HA and NA open reading frames (ORF). Then, the nucleotide sequences from MARMs and no-Ab control groups were determined (ACGT, Inc). Primer sequences for amplification of the pHlNl HA and NA ORFs are available upon request. Virus growth kinetics
  • Multicycle vims growth kinetics were performed in confluent monolayers of MDCK cells (12-well plate format, 5 x 10 5 cells/well, triplicates) infected (MOI 0.001) with the indicated viruses.
  • Virus titers in TCS were determined by immunofocus assay (FFU/ml) (Nogales A, et al. 2014. J Virol 88: 10525-10540 and Nogales A, et al. 2016. J Virol 90:6291- 6302).
  • Mean value and standard deviation (SD) were calculated using MICROSOFT EXCEL software.
  • Hemagglutination inhibition (HAI) assays were used to determine the HA- neutralizing capability of KPF1.
  • the assay was performed as describe previously (Nogales A, et al. 2014. J Virol 88: 10525-10540 and Nogales A, et al. 2016. J Virol 90:6291-6302). Briefly, the KPFlhmAb was serially diluted (2-fold) in 96-well V-bottom plates and mixed 1 : 1 with 4 hemagglutinating units (HAU) of pHlNl for 60 min at room temperature.
  • HAU hemagglutinating units
  • the HAI titers were determined by adding 0.5% turkey red blood cells (RBCs) to the virus-hmAb mixtures for 30 min on ice. The HAI titer was defined as the minimum amount of hmAb that completely inhibited hemagglutination.
  • Peripheral blood plasmablasts (CD19+IgD-CD38+CD27++) were single cell sorted from a healthy subject seven days after immunization with the 2014-2015 seasonal inactivated quadrivalent influenza vaccine. This subject had a robust plasmablast response comprising -25% of the IgD- compartment that were predominantly CD20-CD126(IL-6Ra)+ (Fig. 1A), consistent with an antigen-specific response (Gonzalez-Garcia I, et al. 2006. J Immunol 176:4042-4050).
  • the plasmablasts were subjected to single-cell RT-PCR and sequencing of the immunoglobulin variable regions and the dominant B cell lineage, comprising -10% of the isolated plasmablasts was identified and is encoded by the immunoglobulin (Ig) heavy chain VH3-23 and kappa light chain Vkl-33 genes. Due to the dominance of this lineage among the plasmablasts, a representative member of the lineage was cloned as an IgGl to generate the KPF1 fully hmAb.
  • the heavy chain variable region (VH) contained 14% amino acid (8% NT) and kappa light chain variable region (Vk) 10% amino acids (5% NT) mutations from the germline, consistent with affinity maturation (Fig. IB).
  • this lineage included only IgAl .
  • One constant region-encoding nucleic acid derived from the B cell lineage was aligned against a nucleic acid encoding a wild type IgAl constant region (Fig. 1C).
  • a representative member of the lineage was cloned as an IgGl to generate the KPFl fully hmAb.
  • KPFl bound only to HI HAs including A/South Carolina/01/1918 H1N1 with a trend of greater reactivity to more recent H1N1 strains (Fig. 3).
  • the HI reactivity profile for KPFl mirrored the subject's plasma Ab response, including its strong reactivity against TX H1N1, which dominated the subject's pre-vaccination (DO) plasma response.
  • KPFl hemagglutination inhibition
  • Viruses used in this assay A/California/04/09 HlNl (pHlNl), A/Texas/36/91 HlNl (TX HlNl), A/New Caledonia/20/99 HlNl (NC HlNl), A/Wyoming/3/03 H3N2
  • H3N2 A/Puerto Rico/08/34 HlNl (PR8 HlNl), or B/Brisbane/60/08 (IBV).
  • MDCK cells were infected (100 PFU) with the indicated viruses, which were pre- incubated with 2-fold serial dilutions (starting concentration of 200 ⁇ g/ml) of the hmAb KPF1.
  • starting concentration of 200 ⁇ g/ml 2-fold serial dilutions
  • NT50 2-fold serial dilutions
  • HAI assays were performed using 2-fold serial dilutions (starting concentration of 200 ⁇ g/ml) of KPF1 and 4 hemagglutinating units (HAU) of the indicated viruses.
  • HAI titers were determined by adding 0.5% turkey RBCs to the virus-hmAb mixtures and defined as the minimum amount of hmAb that completely inhibited hemagglutination.
  • mice treated with 1 mg/kg or 10 mg/kg had significant reductions in viral titers in their lungs at two and four days p.i., including the absence of detectable virus in 2 of 3 mice treated with 10 mg/kg, suggestive of sterilizing immunity in these mice (Fig. 5C).
  • mice were treated with 10 mg/kg of IgG isotype control or with KPFl and then challenged with a lethal dose (lOx MLD50) of PR8, TX, or NC H1N1 influenza viruses (Pica, N. et al. J Virol 85, 12825- 12829, https://doi.org/10.1128/JVI.05930-l l (2011); and Nogales, A. et al.
  • mAb-resistant mutants of pHlNl were generated (Fig. 7).
  • WT pHlNl virus was passaged in triplicates for five rounds in the presence of increasing concentrations of KPFl and the NA and HA ORFs were sequenced from three MARMs (MARMl, MARM2, MARM3) (Fig. 7A).
  • MARMl, MARM2, MARM3 No mutations were detected in NA and all three MARMs shared a point mutation (E129K) located between the Ca and Cb antigenic sites.
  • MARM3 also had an additional mutation (K180N), which is located within the Sa antigenic site (Figs. 7A and 7D).
  • KPFl did not bind to MARM1-3 as determined by immunofluorescence, although the binding to an NP-specific mAb and another pHlNl head-specific HA mAb 29E3 was maintained (Fig. 7B).
  • In vitro viral growth kinetics of MARMl and MARM2 were comparable to that of pHlNl WT, although the growth of MARM3 was significantly less (p ⁇ 0.05) than pHlNl WT, suggesting that K180N mutation impacts viral fitness (Fig. 7C).
  • the amino acids in the position 129 and 180 are far away from each other in the linear sequence of the HA protein, the analysis of the tridimensional protein structure shows that both amino acids are close in the folded HA protein (Fig. 7D).
  • E129 is important for the binding of KPFl to HA and suggest that KPFl recognizes an epitope in the HI HA globular head that dependent on residues near the Ca and Cb antigenic sites (E129) (Fig. 7d).
  • pCAGGS protein expression plasmids encoding the WT, single E129K, K180N, K180Q; or double E129K/K180N HA mutants were generated.
  • the ability of KPFl to recognize the different HAs was evaluated by immunofluorescence assay in transfected 293 T cells (Fig. 8a).
  • KPFl was unable to recognize HA proteins containing the amino acid change E129K (E129K and E129K/K180N) but was able to recognize HA proteins containing the amino acid change at position 180 (K180N and K180Q). This suggests that position 180 is not part of the KPFl hmAb footprint and the amino acid change observed in 1 of 3 MARMS at position 180 was probably a random event or a compensatory mutation.
  • position 180 has higher variability, showing a shift from K to Q in the last decade (Fig. 8B). However, this variability does not affect the binding of KPFl to HA.
  • the KPFl antibody has broad activity against HI influenza isolates and potent prophylactic and therapeutic activity in vivo, which is mediated by recognition of conserved residues in the HI hemagglutinin globular head. This antibody, as well as others disclosed herein, have greater clinical feasibility.

Abstract

La présente invention concerne des anticorps monoclonaux anti-virus de la grippe à large neutralisation ou des fragments de liaison à l'antigène de ceux-ci. La présente invention concerne en outre des utilisations thérapeutiques de l'anticorps isolé ou du fragment de liaison à l'antigène de celui-ci.
EP18732970.1A 2017-05-15 2018-05-10 Anticorps monoclonal anti-virus de la grippe à large neutralisation et utilisations associées Pending EP3625251A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762506256P 2017-05-15 2017-05-15
PCT/US2018/032063 WO2018213097A1 (fr) 2017-05-15 2018-05-10 Anticorps monoclonal anti-virus de la grippe à large neutralisation et utilisations associées

Publications (1)

Publication Number Publication Date
EP3625251A1 true EP3625251A1 (fr) 2020-03-25

Family

ID=62685085

Family Applications (1)

Application Number Title Priority Date Filing Date
EP18732970.1A Pending EP3625251A1 (fr) 2017-05-15 2018-05-10 Anticorps monoclonal anti-virus de la grippe à large neutralisation et utilisations associées

Country Status (4)

Country Link
US (1) US11168129B2 (fr)
EP (1) EP3625251A1 (fr)
CN (1) CN111094335B (fr)
WO (1) WO2018213097A1 (fr)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20230107644A1 (en) 2020-04-01 2023-04-06 University Of Rochester Monoclonal antibodies against the hemagglutinin (ha) and neuraminidase (na) of influenza h3n2 viruses

Family Cites Families (87)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4475196A (en) 1981-03-06 1984-10-02 Zor Clair G Instrument for locating faults in aircraft passenger reading light and attendant call control system
US4447233A (en) 1981-04-10 1984-05-08 Parker-Hannifin Corporation Medication infusion pump
US4439196A (en) 1982-03-18 1984-03-27 Merck & Co., Inc. Osmotic drug delivery system
US4522811A (en) 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
US4447224A (en) 1982-09-20 1984-05-08 Infusaid Corporation Variable flow implantable infusion apparatus
US4487603A (en) 1982-11-26 1984-12-11 Cordis Corporation Implantable microinfusion pump system
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4486194A (en) 1983-06-08 1984-12-04 James Ferrara Therapeutic device for administering medicaments through the skin
EP0154316B1 (fr) 1984-03-06 1989-09-13 Takeda Chemical Industries, Ltd. Lymphokine chimiquement modifiée et son procédé de préparation
US4596556A (en) 1985-03-25 1986-06-24 Bioject, Inc. Hypodermic injection apparatus
US5374548A (en) 1986-05-02 1994-12-20 Genentech, Inc. Methods and compositions for the attachment of proteins to liposomes using a glycophospholipid anchor
MX9203291A (es) 1985-06-26 1992-08-01 Liposome Co Inc Metodo para acoplamiento de liposomas.
US6548640B1 (en) 1986-03-27 2003-04-15 Btg International Limited Altered antibodies
DE3883899T3 (de) 1987-03-18 1999-04-22 Sb2 Inc Geänderte antikörper.
US4941880A (en) 1987-06-19 1990-07-17 Bioject, Inc. Pre-filled ampule and non-invasive hypodermic injection device assembly
US4790824A (en) 1987-06-19 1988-12-13 Bioject, Inc. Non-invasive hypodermic injection device
US5677425A (en) 1987-09-04 1997-10-14 Celltech Therapeutics Limited Recombinant antibody
JP2919890B2 (ja) 1988-11-11 1999-07-19 メディカル リサーチ カウンスル 単一ドメインリガンド、そのリガンドからなる受容体、その製造方法、ならびにそのリガンドおよび受容体の使用
EP0401384B1 (fr) 1988-12-22 1996-03-13 Kirin-Amgen, Inc. Facteur de stimulation de colonies de granulocytes modifies chimiquement
US5108921A (en) 1989-04-03 1992-04-28 Purdue Research Foundation Method for enhanced transmembrane transport of exogenous molecules
DE3920358A1 (de) 1989-06-22 1991-01-17 Behringwerke Ag Bispezifische und oligospezifische, mono- und oligovalente antikoerperkonstrukte, ihre herstellung und verwendung
US5959177A (en) 1989-10-27 1999-09-28 The Scripps Research Institute Transgenic plants expressing assembled secretory antibodies
US5312335A (en) 1989-11-09 1994-05-17 Bioject Inc. Needleless hypodermic injection device
US5064413A (en) 1989-11-09 1991-11-12 Bioject, Inc. Needleless hypodermic injection device
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
ATE164395T1 (de) 1990-12-03 1998-04-15 Genentech Inc Verfahren zur anreicherung von proteinvarianten mit geänderten bindungseigenschaften
US5571894A (en) 1991-02-05 1996-11-05 Ciba-Geigy Corporation Recombinant antibodies specific for a growth factor receptor
DK0590058T3 (da) 1991-06-14 2004-03-29 Genentech Inc Humaniseret heregulin-antistof
GB9114948D0 (en) 1991-07-11 1991-08-28 Pfizer Ltd Process for preparing sertraline intermediates
WO1993006217A1 (fr) 1991-09-19 1993-04-01 Genentech, Inc. EXPRESSION DANS L'E. COLI DE FRAGMENTS D'ANTICORPS POSSEDANT AU MOINS UNE CYSTEINE PRESENTE SOUS FORME D'UN THIOL LIBRE, ET LEUR UTILISATION DANS LA PRODUCTION D'ANTICORPS BIFONCTIONNELS F(ab')¿2?
US5587458A (en) 1991-10-07 1996-12-24 Aronex Pharmaceuticals, Inc. Anti-erbB-2 antibodies, combinations thereof, and therapeutic and diagnostic uses thereof
EP1997894B1 (fr) 1992-02-06 2011-03-30 Novartis Vaccines and Diagnostics, Inc. Protéine de liaison biosynthétique pour un marqueur du cancer
US5714350A (en) 1992-03-09 1998-02-03 Protein Design Labs, Inc. Increasing antibody affinity by altering glycosylation in the immunoglobulin variable region
WO1993022332A2 (fr) 1992-04-24 1993-11-11 Board Of Regents, The University Of Texas System Production recombinante de domaines semblables a l'immunoglobuline dans des cellules procaryotes
US5383851A (en) 1992-07-24 1995-01-24 Bioject Inc. Needleless hypodermic injection device
CA2163345A1 (fr) 1993-06-16 1994-12-22 Susan Adrienne Morgan Anticorps
US5789199A (en) 1994-11-03 1998-08-04 Genentech, Inc. Process for bacterial production of polypeptides
US5840523A (en) 1995-03-01 1998-11-24 Genetech, Inc. Methods and compositions for secretion of heterologous polypeptides
US5869046A (en) 1995-04-14 1999-02-09 Genentech, Inc. Altered polypeptides with increased half-life
US6096871A (en) 1995-04-14 2000-08-01 Genentech, Inc. Polypeptides altered to contain an epitope from the Fc region of an IgG molecule for increased half-life
US6121022A (en) 1995-04-14 2000-09-19 Genentech, Inc. Altered polypeptides with increased half-life
JP4046354B2 (ja) 1996-03-18 2008-02-13 ボード オブ リージェンツ,ザ ユニバーシティ オブ テキサス システム 増大した半減期を有する免疫グロブリン様ドメイン
US6277375B1 (en) 1997-03-03 2001-08-21 Board Of Regents, The University Of Texas System Immunoglobulin-like domains with increased half-lives
US6040498A (en) 1998-08-11 2000-03-21 North Caroline State University Genetically engineered duckweed
US6610833B1 (en) 1997-11-24 2003-08-26 The Institute For Human Genetics And Biochemistry Monoclonal human natural antibodies
ES2375931T3 (es) 1997-12-05 2012-03-07 The Scripps Research Institute Humanización de anticuerpo murino.
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
EP2261229A3 (fr) 1998-04-20 2011-03-23 GlycArt Biotechnology AG Modification par glycosylation d'anticorps aux fins d'amélioration de la cytotoxicité cellulaire dépendant des anticorps
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
JP2003512019A (ja) 1999-01-15 2003-04-02 ジェネンテック・インコーポレーテッド 変化したエフェクター機能を有するポリペプチド変異体
ES2420835T3 (es) 1999-04-09 2013-08-27 Kyowa Hakko Kirin Co., Ltd. Procedimiento para controlar la actividad de las moléculas inmunofuncionales
MXPA02003456A (es) 1999-10-04 2002-10-23 Medicago Inc Metodo para regular la transcripcion de genes foraneos.
US7125978B1 (en) 1999-10-04 2006-10-24 Medicago Inc. Promoter for regulating expression of foreign genes
WO2001044463A1 (fr) 1999-12-15 2001-06-21 Genentech, Inc. Balayage aveugle, procede combinatoire permettant la representation d'epitopes de proteines fonctionnelles
CA2399832C (fr) 2000-02-11 2011-09-20 Stephen D. Gillies Amelioration de la demi-vie circulante de proteines de fusion a base d'anticorps
US6725230B2 (en) 2000-07-18 2004-04-20 Aegis Analytical Corporation System, method and computer program for assembling process data of multi-database origins using a hierarchical display
US6596541B2 (en) 2000-10-31 2003-07-22 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
CN101940189A (zh) 2000-11-30 2011-01-12 米德列斯公司 用于生产人类抗体的转基因转染色体啮齿动物
ES2326964T3 (es) 2001-10-25 2009-10-22 Genentech, Inc. Composiciones de glicoproteina.
US20040002587A1 (en) 2002-02-20 2004-01-01 Watkins Jeffry D. Fc region variants
US7317091B2 (en) 2002-03-01 2008-01-08 Xencor, Inc. Optimized Fc variants
US20040132101A1 (en) 2002-09-27 2004-07-08 Xencor Optimized Fc variants and methods for their generation
AU2003239966B9 (en) 2002-06-03 2010-08-26 Genentech, Inc. Synthetic antibody phage libraries
WO2004016750A2 (fr) 2002-08-14 2004-02-26 Macrogenics, Inc. Anticorps specifiques du recepteur fc$g(g)riib et procedes d'utilisation de ces anticorps
DK2345671T3 (en) 2002-09-27 2016-02-15 Xencor Inc Optimized Fc variants and methods for their formation
DK1562972T3 (da) 2002-10-15 2010-12-06 Facet Biotech Corp Modifikation af FcRn-bindingsaffiniteter eller serumhalveringstider for antistoffer ved mutagenese
AU2004204494B2 (en) 2003-01-09 2011-09-29 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
EP1585767A2 (fr) 2003-01-16 2005-10-19 Genentech, Inc. Banques de phages anticorps synthetiques
US8101720B2 (en) 2004-10-21 2012-01-24 Xencor, Inc. Immunoglobulin insertions, deletions and substitutions
GB0324368D0 (en) 2003-10-17 2003-11-19 Univ Cambridge Tech Polypeptides including modified constant regions
EA009746B1 (ru) 2004-01-12 2008-04-28 Эпплайд Молекьюлар Эволюшн, Инк. ВАРИАНТЫ УЧАСТКА Fc
EP2053062A1 (fr) 2004-03-24 2009-04-29 Xencor, Inc. Variantes d'immunoglobine en dehors de la région Fc
JP5128935B2 (ja) 2004-03-31 2013-01-23 ジェネンテック, インコーポレイテッド ヒト化抗TGF−β抗体
US7785903B2 (en) 2004-04-09 2010-08-31 Genentech, Inc. Variable domain library and uses
EA012464B1 (ru) 2004-08-04 2009-10-30 Эпплайд Молекьюлар Эволюшн, Инк. Антитело против cd20 и его применение
EP1957531B1 (fr) 2005-11-07 2016-04-13 Genentech, Inc. Polypeptides de liaison comprenant des sequences diversifiees et des sequences consensus hypervariables vh/vl
EP1973951A2 (fr) 2005-12-02 2008-10-01 Genentech, Inc. Polypeptides de liaison avec des sequences de diversite limitees
TW200812616A (en) 2006-05-09 2008-03-16 Genentech Inc Binding polypeptides with optimized scaffolds
CN100592373C (zh) 2007-05-25 2010-02-24 群康科技(深圳)有限公司 液晶显示面板驱动装置及其驱动方法
CN103665156B (zh) * 2012-09-14 2016-08-03 中国科学院上海生命科学研究院 一种抗流感病毒广谱中和性的结合分子3e1
AR093446A1 (es) * 2012-11-13 2015-06-10 Genentech Inc Anticuerpos de antihemaglutinina y metodos de uso
ES2682981T3 (es) 2013-08-03 2018-09-24 Avatar Medical, Llc Proteínas de hemaglutinina del virus de la gripe y método de uso de las mismas
US9975925B2 (en) 2013-08-28 2018-05-22 Glaxosmithkline Biologicals S.A. Influenza antigens and antibodies
US10435462B2 (en) * 2015-03-31 2019-10-08 Viro Dynamics Corporation Hybrid influenza seed viruses, compositions thereof, and use thereof in the diagnosis or therapy of influenza
WO2016205347A1 (fr) * 2015-06-16 2016-12-22 Icahn School Of Medicine At Mount Sinai Vaccins contre le virus de la grippe et leurs utilisations

Also Published As

Publication number Publication date
US11168129B2 (en) 2021-11-09
CN111094335A (zh) 2020-05-01
US20200140526A1 (en) 2020-05-07
CN111094335B (zh) 2022-08-23
WO2018213097A1 (fr) 2018-11-22

Similar Documents

Publication Publication Date Title
US10815294B2 (en) Neutralizing anti-influenza A virus antibodies and uses thereof
US11634477B2 (en) Neutralizing anti-SARS-CoV-2 antibodies and methods of use thereof
US10882897B2 (en) Neutralizing anti-influenza binding molecules and uses thereof
JP7402541B2 (ja) 抗インフルエンザノイラミニダーゼモノクローナル抗体およびその使用
US11168129B2 (en) Broadly neutralizing anti-influenza human monoclonal antibody and uses thereof
US11958895B2 (en) Anti-influenza neuraminidase monoclonal antibodies and uses thereof
US20230107644A1 (en) Monoclonal antibodies against the hemagglutinin (ha) and neuraminidase (na) of influenza h3n2 viruses
CA3200048A1 (fr) Anticorps largement neutralisants diriges contre l'encephalite a tiques et virus apparentes

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20191126

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20210309

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230519